EP3143046A1 - Protéines de liaison à l'antigène her1 se liant à l'épingle à cheveux beta de her1 - Google Patents

Protéines de liaison à l'antigène her1 se liant à l'épingle à cheveux beta de her1

Info

Publication number
EP3143046A1
EP3143046A1 EP15721718.3A EP15721718A EP3143046A1 EP 3143046 A1 EP3143046 A1 EP 3143046A1 EP 15721718 A EP15721718 A EP 15721718A EP 3143046 A1 EP3143046 A1 EP 3143046A1
Authority
EP
European Patent Office
Prior art keywords
antibody
herl
seq
human
her1
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP15721718.3A
Other languages
German (de)
English (en)
Other versions
EP3143046B1 (fr
Inventor
Birgit Bossenmaier
Michael Gerg
Gerhard Niederfellner
Michael Schraeml
Carmen PEESS
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
F Hoffmann La Roche AG
Original Assignee
F Hoffmann La Roche AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by F Hoffmann La Roche AG filed Critical F Hoffmann La Roche AG
Publication of EP3143046A1 publication Critical patent/EP3143046A1/fr
Application granted granted Critical
Publication of EP3143046B1 publication Critical patent/EP3143046B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3076Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6068Other bacterial proteins, e.g. OMP
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell

Definitions

  • the invention relates to anti-HERl antigen binding proteins, e.g. anti-HERl antibodies, that bind to the beta-hairpin of HER1, methods for selecting these antigen binding proteins, their preparation and use as medicament.
  • anti-HERl antigen binding proteins e.g. anti-HERl antibodies
  • the HER protein family consists of 4 members: HER1, also named epidermal growth factor receptor (EGFR) or ErbB-1, HER2, also named ErbB-2, ErbB-3, also named HER3 and ErbB-4, also named HER4.
  • the ErbB family proteins are receptor tyrosine kinases and represent important mediators of cell growth, differentiation and survival.
  • the HER family represent receptor proteins of different ligands of the epidermal growth factor family (EGF-family) like epidermal growth factor (EGF), the neuregulin (NRG) family, amphiregulin, and transforming growth factor-a (TGF-a).
  • Human HER1 (also known as epidermal growth factor receptor EGFR or Erb-Bl,) is a 170 kDa transmembrane receptor encoded by the c-erbB proto-oncogene, and exhibits intrinsic tyrosine kinase activity (Modjtahedi, H., et al., Br. J. Cancer 73 (1996) 228-235; Herbst, R.S., and Shin, D.M., Cancer 94 (2002) 1593-1611).
  • SwissProt database entry P00533 provides the sequence of HER1 (SEQ ID NO: 2).
  • HER1 e.g., alternative RNA transcripts, truncated versions, polymorphisms, etc.
  • isoforms and variants of HER1 including but not limited to those identified by Swissprot database entry numbers P00533-1, P00533-2, P00533-3, and P00533-4.
  • HER1 is known to bind ligands including a), epidermal growth factor (EGF), transforming growth factor-a (TGF), amphiregulin, heparin-binding EGF (hb-EGF), betacellulin, and epiregulin (Herbst, R.S., and Shin, D.M., Cancer 94 (2002) 1593-1611; Mendelsohn, J., and Baselga, J., Oncogene 19 (2000) 6550- 6565).
  • EGF epidermal growth factor
  • TGF transforming growth factor-a
  • hb-EGF heparin-binding EGF
  • betacellulin betacellulin
  • epiregulin Herbst, R.S., and Shin, D.M., Cancer 94 (2002) 1593-1611; Mendelsohn, J., and Baselga, J., Oncogene 19 (2000) 6550- 6565.
  • HER1 regulates numerous cellular processes via tyrosine-kinase mediated signal transduction pathways, including, but not limited to, activation of signal transduction pathways that control cell proliferation, differentiation, cell survival, apoptosis, angiogenesis, mitogenesis, and metastasis (Atalay, G., et al, Ann. Oncology 14 (2003) 1346-1363; Tsao, A.S., and Herbst, R.S., Signal 4 (2003) 4-9;
  • Unconjugated monoclonal antibodies can be useful medicines for the treatment of cancer, as demonstrated by the U.S. Food and Drug Administration's approval of Trastuzumab (HerceptinTM; Genentech Inc,) for the treatment of advanced breast cancer (Grillo-Lopez, A.J., et al, Semin. Oncol. 26 (1999) 66-73; Goldenberg, M.M., Clin. Ther. 21 (1999) 309-18), Rituximab (RituxanTM; IDEC
  • Monoclonal antibodies target the extracellular portion of EGFR, which results in blocking ligand binding and thereby inhibits downstream events such as cell proliferation (Tsao, A.S., and Herbst, R.S., Signal 4 (2003) 4-9).
  • EMD 55900 is a murine anti-EGFR monoclonal antibody that was raised against human epidermoid carcinoma cell line A431 and was tested in clinical studies of patients with advanced squamous cell carcinoma of the larynx or hypopharynx (Bier, H., et al., Eur. Arch. Otohinolaryngol. 252 (1995) 433-9).
  • the rat monoclonal antibodies ICR16, ICR62, and ICR80 which bind the extracellular domain of EGFR, have been shown to be effective at inhibiting the binding of EGF and TGF-a the receptor.
  • the murine monoclonal antibody 425 is another MAb that was raised against the human A431 carcinoma cell line and was found to bind to a polypeptide epitope on the external domain of the human epidermal growth factor receptor. (Murthy, U., et al, Arch. Biochem. Biophys. 252(2) (1987) 549-560.
  • a potential problem with the use of murine antibodies in therapeutic treatments is that non-human monoclonal antibodies can be recognized by the human host as a foreign protein; therefore, repeated injections of such foreign antibodies can lead to the induction of immune responses leading to harmful hypersensitivity reactions.
  • murine-based monoclonal antibodies this is often referred to as a Human Anti- Mouse Antibody response, or "HAMA” response, or a Human Anti-Rat Antibody, or "HARA” response.
  • HAMA Human Anti- Mouse Antibody response
  • HSA Human Anti-Rat Antibody
  • these "foreign” antibodies can be attacked by the immune system of the host such that they are, in effect, neutralized before they reach their target site.
  • non-human monoclonal antibodies typically lack human effector functionality, i.e., they are unable to, inter alia, mediate complement dependent lysis or lyse human target cells through antibody dependent cellular toxicity or Fc-receptor mediated phagocytosis.
  • Chimeric antibodies comprising portions of antibodies from two or more different species (e.g., mouse and human) have been developed as an alternative to "conjugated" antibodies.
  • US 5,891,996 (Mateo de Acosta del Rio, CM., et al.) discusses a mouse/human chimeric antibody, R3, directed against
  • EGFR and US 5,558,864 discusses generation of chimeric and humanized forms of the murine anti-EGFR MAb 425.
  • IMC-C225 (Erbitux®; ImClone) is a chimeric mouse/human anti-EGFR monoclonal antibody (based on mouse M225 monoclonal antibody, which resulted in HAMA responses in human clinical trials) that has been reported to demonstrate antitumor efficacy in various human xenograft models. (Herbst, R.S., and Shin, D.M., Cancer 94 (2002) 1593-161 1).
  • IMC-C225 has been attributed to several mechanisms, including inhibition of cell events regulated by EGFR signaling pathways, and possibly by increased antibody-dependent cellular toxicity (ADCC) activity (Herbst, R.S., and Shin, D.M., Cancer 94 (2002) 1593-161 1). IMC-C225 was also used in clinical trials, including in combination with radiotherapy and chemotherapy (Herbst, R.S., and Shin, D.M., Cancer 94 (2002) 1593-161 1). Recently, Abgenix, Inc. (Fremont, CA) developed ABX-EGF for cancer therapy. ABX-EGF is a fully human anti- EGFR monoclonal antibody.
  • the invention provides a method for selecting an antigen binding protein, in particular an antibody, that binds to human HER1 , wherein the antigen binding protein, in particular the antibody, binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NO: l) of human HER1 ; wherein a) at least one polypeptide selected from the group consisting of:
  • SEQ ID NO: 18 TgSlyDcys-HERl which comprises the amino acid sequence of SEQ ID NO:l; is used to select antigen binding proteins, in particular antibodies, which show binding to the at least one polypeptide under a), and thereby selecting an antigen binding protein, in particular an antibody that binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NO : 1 ) of human HER1.
  • the invention provides an antigen binding protein, in particular an antibody, obtained by such selection method.
  • the invention provides an isolated an antigen binding protein, in particular an antibody, that binds to human HER1, wherein the antigen binding protein, in particular the antibody, binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NOT) of human HER1.
  • the invention further provides an isolated antigen binding protein that binds to human HER1, wherein the antigen binding protein binds to a polypeptide of SEQ ID NO: 13 TtSlyDcys-HERl; or
  • SEQ ID NO: 18 TgSlyDcys-HERl .
  • the invention further provides an isolated antigen binding protein that binds to human HER1, wherein the antigen binding protein binds to a polypeptide of SEQ ID NO: 13 TtSlyDcys-HERl .
  • the invention further provides an isolated antigen binding protein that binds to human HER1, wherein the antigen binding protein binds to a polypeptide of
  • the invention further provides an isolated antigen binding protein that binds to human HER1 , wherein the antigen binding protein binds within an amino acid sequence of
  • PPLMLYNPTTYQMDVNPEGK (SEQ ID NOT) which is comprised in a polypeptide of SEQ ID NO: 13 TtSlyDcys-HERl .
  • the invention further provides an isolated antigen binding protein that binds to human HER1 , wherein the antigen binding protein binds within an amino acid sequence of
  • PPLMLYNPTTYQMDVNPEGK (SEQ ID NOT) which is comprised in a polypeptide of SEQ ID NO: 18 TgSlyDcys-HERl .
  • the invention further provides an isolated antibody that binds to human HER1 , wherein the antibody binds to a polypeptide of
  • SEQ ID NO: 18 TgSlyDcys-HERl .
  • the invention further provides an isolated antibody that binds to human HER1 , wherein the antibody binds to a polypeptide of
  • the invention further provides an isolated antibody that binds to human HER1 , wherein the antibody binds to a polypeptide of
  • SEQ ID NO: 18 TgSlyDcys-HERl .
  • the invention further provides an isolated antibody that binds to human HER1 , wherein the antibody binds within an amino acid sequence of
  • PPLMLYNPTTYQMDVNPEGK (SEQ ID NOT) which is comprised in a polypeptide of SEQ ID NO: 13 TtSlyDcys-HERl .
  • the invention further provides an isolated antibody that binds to human HER1 , wherein the antibody binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NOT) which is comprised in a polypeptide of SEQ ID NO: 18 TgSlyDcys-HERl .
  • the isolated antigen binding protein or antibody does not induce phosphorylation of HERl in A549 cancer cells (ATCC CCL-185) in the absence of EGF.
  • the isolated antigen binding protein or antibody is non-agonistic with respect to the phosphorylation of HERl in A549 cancer cells
  • the invention provides an isolated antibody that binds to human HERl , wherein the antibody binds to the amino acid sequence SEQ ID NO: l in activated HERl .
  • the invention provides an isolated antibody that binds to human HERl , wherein the antibody binds to the amino acid sequence SEQ ID NO: l in activated HERl ; and inhibits the homodimerisation of HERl homodimers.
  • the invention provides an isolated antibody that binds to human HERl , wherein the antibody binds to the amino acid sequence SEQ ID NO: l in activated HERl ; and inhibits the heterodimerisation of HER1/HER2 heterodimers.
  • the invention provides an isolated antibody that binds to human HERl , wherein the antibody has on or more of the following properties: a) binds to the amino acid sequence of SEQ ID NO: 1 ; and/or b) binds to the amino acid sequence SEQ ID NO: l in activated HERl ; and/or c) binds within an amino acid sequence of
  • PPLMLYNPTTYQMDVNPEGK (SEQ ID NO: l) which is comprised in a polypeptide selected from the group consisting of:
  • the antibody binds to a polypeptide with a length of 15 amino acids comprising the amino acid sequence TYQMDVNPEG (SEQ ID NO: 19); and /or h) binds to a polypeptide consisting of TYQMDVNPEG (SEQ ID NO: 19); and/or i) the antibody binds to a polypeptide with a length of 15 amino acids comprising the amino acid sequence MLYNPTTYQ (SEQ ID NO:20); and /or j) binds to a polypeptide consisting of MLYNPTTYQ (SEQ ID NO:20); and/or k) does not induce phosphorylation of HER1 in A549 cancer cells (
  • such anti-HERl antibody is a monoclonal antibody.
  • such anti-HERl antibody is a human, humanized, or chimeric antibody.
  • such anti-HERl antibody is an antibody fragment that binds human HER1.
  • One embodiment of the invention is humanized anti-HERl antibody which comprises a) all three heavy chain HVRs and all three light chain HVRs of the deposited antibody MAK ⁇ HER1-DIB> M-50.097.14 (DSM ACC3240); b) all three heavy chain HVRs and all three light chain HVRs of the deposited antibody MAK ⁇ HER1-DIB>M-50.110.23(DSM ACC3241); c) all three heavy chain HVRs and all three light chain HVRs of the deposited antibody MAK ⁇ HER1-DIB> M-37.058.09 (DSM ACC3238); d) all three heavy chain HVRs and all three light chain HVRs of the deposited antibody MAK ⁇ HER1-DIB> M-37.186.15 (DSM ACC3239).
  • anti-HERl antibody is a full length IgGl antibody or IgG4 antibody.
  • such anti-HERl antibody is a Fab fragment.
  • the invention further provides an isolated nucleic acid such anti-HERl antibody.
  • the invention further provides a host cell comprising such nucleic acid.
  • the invention further provides a method of producing an antibody comprising culturing such host cell so that the antibody is produced.
  • such method further comprises recovering the antibody from the host cell.
  • the invention further provides an immunoconjugate comprising such anti-HERl antibody and a cytotoxic agent.
  • the invention further provides a pharmaceutical formulation comprising such anti- HERl antibody and a pharmaceutically acceptable carrier.
  • the invention further provides the anti-HERl antibody described herein for use as a medicament.
  • the invention further provides the anti-HERl antibody described herein, or the immunoconjugate comprising the anti-HERl antibody and a cytotoxic agent, for use in treating cancer.
  • the invention further provides the anti- HERl antibody described herein for use in inhibition of HER family dimerization (HERl homo- or heterodimerization, e.g. HERl homodimerization, or HER1/HER2 heterodimerization)
  • Use of such anti-HERl antibody, or an immunoconjugate comprising the anti- HERl antibody and a cytotoxic agent, in the manufacture of a medicament Such use wherein the medicament is for treatment of cancer.
  • Such use wherein the medicament is for the inhibition of HERl homo- or heterodimerization.
  • the invention further provides a method of treating an individual having cancer comprising administering to the individual an effective amount of the anti-HERl antibody described herein, or an immunoconjugate comprising the anti-HERl antibody and a cytotoxic agent.
  • the invention further provides a method of inducing apoptosis in a cancer cell in an individual suffering from cancer comprising administering to the individual an effective amount of an immunoconjugate comprising the anti-HERl antibody described herein and a cytotoxic agent, thereby inducing apoptosis in a cancer cell in the individual.
  • One embodiment of the invention is a polypeptide selected from the group consisting of: i) SEQ ID NO 12 TtSlyDcas-HERl,
  • SEQ ID NO 18 TgSlyDcys-HERl, which polypeptide comprises the amino acid sequence of SEQ ID NO: 1.
  • the invention further provides the use of one of such polypeptides selected from the group consisting of: i) SEQ ID NO 12 TtSlyDcas-HERl,
  • the invention further provides a method for producing an antibody specifically binding to the B-hairpin of HERl with the amino acid sequence of SEQ ID NO: l comprising the following steps: a) administering to an experimental animal a polypeptide selected from the group consisting of: i) SEQ ID NO 12 TtSlyDcas-HERl,
  • the invention further provides the use of a polypeptide selected from the group consisting of: i) SEQ ID NO 12 TtSlyDcas-HERl,
  • SEQ ID NO 18 TgSlyDcys-HERl, for epitope mapping, whereby the polypeptide comprises the epitope in the B- hairpin of HERl with the amino acid sequence of SEQ ID NO: 1.
  • the anti-HERl antigen binding proteins, in particular antibodies, described herein binding to this beta-hairpin could be selected. It was found that the antigen binding proteins, in particular antibodies, according to the invention have highly valuable properties such as the binding to the activated form of HERl , so they show a clearly stronger HERl internalization in the presence than in the absence of EGF.
  • Figure 1 Schematic overview of "closed” and “open” HERl conformation and the influence of the EGF ligand on the conformation change.
  • Figure 2 3D-structure of the beta-hairpin of HERl functionally presented in a 3-dimensional orientation within a SlyD scaffold of Thermus thermophiles.
  • Figure 6 Strategy of the epitope mapping and alanine-scan approach.
  • the peptide hairpin sequences (peptide hairpin) of HERl (EGFR) ECD, Her-2 ECD, Her-3 ECD and Her-4 ECD including their structural embeddings (structural) were investigated. Cysteins were replaced by serines.
  • FIG. 7 CelluSpotsTM Synthesis and Epitope Mapping of epitopes of HERl antibodies M-31-22, M-47-13, M-50-14 (deposited MAK ⁇ HER1- DIB> M-50.097.14) and M-50-23 (deposited MAK ⁇ HER1- DIB>M-50.110.23).
  • M-47-13 binds to HERl ECD binding epitope TYQMDVNPEG (SEQ ID NO: 19).
  • M-50-14 deposited MAK ⁇ HER1-DIB> M-50.097.14
  • M-50-23 deposited MAK ⁇ HERl-DIB>M-50.110.23
  • HERl ECD binding epitope MLYNPTTYQ SEQ ID NO:20
  • Figure 8 8A (A549 cells) and 8B (A431 cells, strong HERl expression with slight constitutively activated/phosphorylated HERl). Left lane shows HERl detection, right lane the phosphorylated HERl detection in the absence or presence of EGF
  • Figure 9 Binding of anti-HERl ⁇ -hairpin antibodies to and internalization of anti-HERl ⁇ -hairpin antibodies was analyzed in Western Blot using the HER1 expressing cancer cell line A549. Left side shows time dependent HER1 detection in the absence of EGF, right side shows time dependent HER1 detection in the presence of EGF. Antibody M-50-14 shows a clearly stronger HER1 internalization in the presence than in the absence of EGF.
  • acceptor human framework for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below.
  • An acceptor human framework "derived from” a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less,
  • the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
  • An "affinity matured” antibody refers to an antibody with one or more alterations in one or more hypervariable regions (HVRs), compared to a parent antibody which does not possess such alterations, such alterations resulting in an improvement in the affinity of the antibody for antigen.
  • the term "antigen binding protein” as used herein refers to an antibody as described herein or to a scaffold antigen binding protein. In one preferred embodiment the antigen binding protein is an antibody as described herein.
  • Scaffold antigen binding proteins are known in the art, for example, fibronectin and designed ankyrin-repeat proteins (DARPins) have been used as alternative scaffolds for antigen-binding domains, see, e.g., Gebauer and Skerra, Engineered protein scaffolds as next-generation antibody therapeutics. Curr Opin Chem Biol 13:245-255 (2009) and Stumpp et al, Darpins: A new generation of protein therapeutics. Drug Discov Today 13: 695-701 (2008), both of which are incorporated herein by reference in their entirety. B. Criteria for Selecting Parent Variable Domains and Receptors for antigen binding proteins of the invention.
  • a scaffold antigen binding protein is selected from the group consisting of CTLA-4 (Evibody); lipocalin; Protein A derived molecules such as Z- domain of Protein A (Affibody, SpA), A-domain (Avimer/Maxibody); Heat shock proteins such as GroEI and GroES; transferrin (trans-body); ankyrin repeat protein (DARPin); peptide aptamer; C-type lectin domain (Tetranectin); human . gamma.
  • CTLA-4 Cytotoxic T Lymphocyte-associated Antigen 4
  • PDZ domains PDZ domains
  • scorpion toxinkunitz type domains of human protease inhibitors PDZ domains
  • fibronectin adnectin
  • Lipocalins are a family of extracellular proteins which transport small hydrophobic molecules such as steroids, bilins, retinoids and lipids. They have a rigid .beta.- sheet secondary structure with a number of loops at the open end of the conical structure which can be engineered to bind to different target antigens. Anticalins are between 160-180 amino acids in size, and are derived from lipocalins. For further details see Biochim Biophys Acta 1482: 337-350 (2000), US7250297B1 and US20070224633.
  • An affibody is a scaffold derived from Protein A of Staphylococcus aureus which can be engineered to bind to antigen.
  • the domain consists of a three-helical bundle of approximately 58 amino acids. Libraries have been generated by randomisation of surface residues.
  • Protein Eng. Des. Sel. 17, 455-462 (2004) and EP1641818AlAvimers are multidomain proteins derived from the A- domain scaffold family. The native domains of approximately 35 amino acids adopt a defined disulphide bonded structure. Diversity is generated by shuffling of the natural variation exhibited by the family of A-domains. For further details see Nature Biotechnology 23(12), 1556 - 1561 (2005) and Expert Opinion on Investigational Drugs 16(6), 909-917 (June 2007).
  • a transferrin is a monomeric serum transport glycoprotein. Transferrins can be engineered to bind different target antigens by insertion of peptide sequences in a permissive surface loop. Examples of engineered transferrin scaffolds include the Trans-body. For further details see J. Biol. Chem 274, 24066-24073 (1999).
  • DARPins Designed Ankyrin Repeat Proteins
  • Ankyrin which is a family of proteins that mediate attachment of integral membrane proteins to the cytoskeleton.
  • a single ankyrin repeat is a 33 residue motif consisting of two
  • .alpha.-helices and a .beta.-turn They can be engineered to bind different target antigens by randomising residues in the first .alpha. -helix and a .beta.-turn of each repeat. Their binding interface can be increased by increasing the number of modules (a method of affinity maturation). For further details see J. MoT Biol. 332, 489-503 (2003), PNAS 100(4), 1700-1705 (2003) and J. MoT Biol. 369, 1015-
  • Fibronectin is a scaffold which can be engineered to bind to antigen.
  • Adnectins consists of a backbone of the natural amino acid sequence of the 10th domain of the 15 repeating units of human fibronectin type III (FN3). Three loops at one end of the .beta. -sandwich can be engineered to enable an Adnectin to specifically recognize a therapeutic target of interest. For further details see Protein Eng. Des. Sel. 18, 435- 444 (2005), US20080139791, WO2005056764 and US6818418B1.
  • Peptide aptamers are combinatorial recognition molecules that consist of a constant scaffold protein, typically thioredoxin (TrxA) which contains a constrained variable peptide loop inserted at the active site.
  • TrxA thioredoxin
  • Microbodies are derived from naturally occurring microproteins of 25-50 amino acids in length which contain 3-4 cysteine bridges - examples of microproteins include KalataBI and conotoxin and knottins.
  • the microproteins have a loop which can be engineered to include up to 25 amino acids without affecting the overall fold of the microprotein.
  • engineered knottin domains see WO2008098796.
  • antigen binding proteins include proteins which have been used as a scaffold to engineer different target antigen binding properties include human . gamma. - crystallin and human ubiquitin (affilins), kunitz type domains of human protease inhibitors, PDZ- domains of the Ras-binding protein AF-6, scorpion toxins (charybdotoxin), C-type lectin domain (tetranectins) are reviewed in Chapter 7 - Non- Antibody Scaffolds from Handbook of Therapeutic Antibodies (2007, edited by Stefan Dubel) and Protein Science 15:14-27 (2006). Epitope binding domains of the present invention could be derived from any of these alternative protein domains.
  • anti-HERl antigen binding protein an antigen binding protein that binds to (human) HERl
  • an antigen binding protein that binds specifically to human HERl refer to an antigen binding protein that is capable of binding
  • the extent of binding of an anti-HERl antigen binding protein to an unrelated, non- HER1 protein is less than about 10% of the binding of the antigen binding protein to HERl as measured, e.g., by a Surface Plasmon Resonance assay (e.g.
  • an antigen binding protein that binds to human HERl has a KD value of the binding affinity for binding to human HERl of ⁇ 1 ⁇ , ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g. 10 " 8 M or less, e.g. from 10 "8 M to 10 "13 M, e.g., from 10 "9 M to 10 "13 M).
  • the respective KD value of the binding affinities is determined in a Surface Plasmon Resonance assay using the wildtype Extracellular domain (ECD) of human HERl (HER1-ECD) for the HERl binding affinity.
  • anti-HERl antibody an antibody that binds to (human) HERl
  • an antibody that binds specifically to human HERl refer to an antibody that is capable of binding HER1 with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting HER1.
  • the extent of binding of an anti-HERl antibody to an unrelated, non-HERl protein is less than about 10% of the binding of the antibody to HER1 as measured, e.g., by a Surface Plasmon Resonance assay (e.g. BIACORE).
  • an antibody that binds to human FIERI has a KD value of the binding affinity for binding to human HER1 of ⁇ 1 ⁇ , ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g. 10 "8 M or less, e.g. from 10 "8 M to 10 "13 M, e.g., from 10 "9 M to 10 "13 M).
  • the respective KD value of the binding affinities is determined in a Surface Plasmon Resonance assay using the wildtype
  • Extracellular domain (ECD) of human HER1 (HER1-ECD) for the HER1 binding affinity.
  • anti-HERl antigen binding protein or anti-HERl antibody that binds to the amino acid sequence SEQ ID NO: l in activated HER1 refers to an anti-HERl antigen binding protein or anti-HERl antibody that binds to the amino acid sequence SEQ ID NO: l comprised in the human HER1-ECD.
  • anti-HERl antigen binding protein or anti- HERl antibody that binds to the amino acid sequence SEQ ID NO: l refers to an anti-HERl antigen binding protein or anti-HERl antibody that binds to the amino acid sequence SEQ ID NO: l comprised in the polypeptide of SEQ ID NO: 13 (TtSlyDcys-HERl).
  • anti-HERl antigen binding protein or anti-HERl antibody that binds to the amino acid sequence SEQ ID NO: l in activated HER1 refers to an anti-HERl antigen binding protein or anti-HERl antibody that binds to the amino acid sequence SEQ ID NO: l comprised in the polypeptide of SEQ ID NO: l
  • antibody herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
  • antibody fragment refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
  • an "antibody that binds to the same epitope” as a reference antibody refers to an antibody that blocks binding of the reference antibody to its antigen in a competition assay by 50% or more, and conversely, the reference antibody blocks binding of the antibody to its antigen in a competition assay by 50% or more.
  • An exemplary competition assay is provided herein.
  • antibody or antigen binding protein that has/shows crossreactivity to (or alternatively that crossreacts with) (human) HER2, HER3 and/or HER4, when it does not crossreacts with Extracellular domain (ECD) of ) human HER2, HER3 and/or HER4, i.e. when the binding signal (in Relative Units (RU)) measured in a Surface Plasmon Resonance assay is below three times the background signal (noise) (e.g at 25 °C with immobilized (for example captured) antibody to which the human HER2, HER3 and/or HER4-ECD as antigen is injected as soluble analyte).
  • RU Relative Units
  • cancer as used herein may be, for example, lung cancer, non small cell lung (NSCL) cancer, bronchioloalviolar cell lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, gastric cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, meso
  • such cancer is a breast cancer, ovarian cancer, cervical cancer, lung cancer or prostate cancer.
  • such cancers are further characterized by HER1 expression or overexpression.
  • the invention are the anti-HERl antibodies of the present invention for use in the simultaneous treatment of primary tumors and new metastases.
  • chimeric antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species.
  • the "class" of an antibody refers to the type of constant domain or constant region possessed by its heavy chain.
  • the heavy chain constant domains that correspond to the different classes of immunoglobulins are called a, ⁇ , ⁇ , ⁇ , and ⁇ respectively.
  • cytotoxic agent refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction. Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At211, 1131, 1125,
  • chemotherapeutic agents or drugs e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents
  • growth inhibitory agents e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents
  • growth inhibitory agents e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents
  • deposited antibody refers to the antibody produced by the respective deposited hybridoma cells identified by the designation and deposition number;. See als o Deposit of biological material below.
  • Effective functions refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor); and B cell activation.
  • An "effective amount” of an agent, e.g., a pharmaceutical formulation refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • epitope includes any polypeptide determinant capable of specific binding to an antibody.
  • epitope determinant include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three dimensional structural characteristics, and or specific charge characteristics.
  • An epitope is a region of an antigen that is bound by an antibody.
  • Fc region herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes native sequence Fc regions and variant Fc regions.
  • a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc region may or may not be present.
  • numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat, E.A. et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991), NIH Publication 91 -3242.
  • FR Framework or "FR” refers to variable domain residues other than hypervariable region (HVR) residues.
  • the FR of a variable domain generally consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FR1-H1(L1)-FR2- H2(L2)-FR3-H3(L3)-FR4.
  • full length antibody is used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
  • host cell refers to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
  • a "human antibody” is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human or a human cell or derived from a non-human source that utilizes human antibody repertoires or other human antibody-encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
  • a "human consensus framework” is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin
  • VL or VH framework sequences are selected from a subgroup of variable domain sequences.
  • the subgroup of sequences is a subgroup as in Kabat, E.A. et al, Sequences of Proteins of Immunological Interest, 5th ed., Bethesda MD (1991), NIH Publication 91-3242, Vols. 1-3.
  • the subgroup is subgroup kappa I as in Kabat et al, supra.
  • the subgroup III is subgroup III as in Kabat et al, supra.
  • a “humanized” antibody refers to a chimeric antibody comprising amino acid residues from non-human HVRs and amino acid residues from human FRs.
  • a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the HVRs (e.g., CDRs) correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody.
  • a humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody.
  • a "humanized variant" of an antibody, e.g., a non- human antibody refers to an antibody that has undergone humanization.
  • a murine HVR is grafted into the framework region of a human antibody to prepare the "humanized antibody.” See e.g. Riechmann, L., et al, Nature 332 (1988) 323-327; and Neuberger, M.S., et al, Nature 314 (1985) 268-270.
  • the murine variable region amino acid sequence is aligned to a collection of human germline antibody V-genes, and sorted according to sequence identity and homology.
  • the acceptor sequence is selected based on high overall sequence homology and optionally also the presence of the right canonical residues already in the acceptor sequence (see Poul, M-A.
  • the germline V-gene encodes only the region up to the beginning of HVR3 for the heavy chain, and till the middle of HVR3 of the light chain. Therefore, the genes of the germline V-genes are not aligned over the whole V-domain.
  • the humanized construct comprises the human frameworks 1 to 3, the murine HVRs, and the human framework 4 sequence derived from the human JK4, and the JH4 sequences for light and heavy chain, respectively.
  • the so-called canonical loop structures of the donor antibody can be determined (see Morea, V., et al, Methods, Vol 20, Issue 3 (2000) 267-279). These canonical loop structures are determined by the type of residues present at the so-called canonical positions. These positions lie (partially) outside of the HVR regions, and should be kept functionally equivalent in the final construct in order to retain the HVR conformation of the parental (donor) antibody.
  • a method for humanizing antibodies comprises the steps of identifying the canonical HVR structure types of the HVRs in a non- human mature antibody; obtaining a library of peptide sequence for human antibody variable regions; determining the canonical HVR structure types of the variable regions in the library; and selecting the human sequences in which the canonical HVR structure is the same as the non-human antibody canonical HVR structure type at corresponding locations within the non-human and human variable regions.
  • the potential acceptor sequence is selected based on high overall homology and optionally in addition the presence of the right canonical residues already in the acceptor sequence. In some cases simple HVR grafting only result in partial retention of the binding specificity of the non-human antibody.
  • non-human framework residues are required for reconstituting the binding specificity and have also to be grafted into the human framework, i.e. so called "back mutations" have to be made in addition to the introduction of the non-human HVRs (see e.g. Queen et al., Proc. Natl. Acad. Sci. USA 86 (1989) 10,029-10,033; Co et al, Nature 351 (1991) 501-502).
  • These specific framework amino acid residues participate in FR-HVR interactions and stabilized the conformation (loop) of the HVRs (see e.g. Kabat et al., J. Immunol.
  • humanized variant of an antibody according to the invention refers to an antibody, which is based on the mouse antibody sequences in which the VH and VL are humanized by above described standard techniques (including HVR grafting and optionally subsequent mutagenesis of certain amino acids in the framework region and the HVR-H1, HVR-H2, HVR-L1 or HVR-L2, whereas HVR-H3 and HVR-L3 remain unmodified).
  • hypervariable region refers to each of the regions of an antibody variable domain which are hypervariable in sequence ("complementarity determining regions” or “CDRs") and/or form structurally defined loops ("hypervariable loops") and/or contain the antigen-contacting residues ("antigen contacts").
  • CDRs complementarity determining regions
  • hypervariable loops form structurally defined loops
  • antigen contacts Generally, antibodies comprise six HVRs: three in the VH (HI, H2, H3), and three in the VL (LI, L2, L3).
  • Exemplary HVRs herein include: (a) hypervariable loops occurring at amino acid residues 26-32 (LI), 50-52 (L2),
  • HVR residues and other residues in the variable domain are numbered herein according to Kabat et al, supra.
  • the HVRs refer to CDRs occurring at amino acid residues 24-34 (LI),
  • HVRs are determined according to Kabat.
  • An "immunoconjugate” is an antibody conjugated to one or more heterologous molecule(s), including but not limited to a cytotoxic agent.
  • An "individual” or “subject” is a mammal.
  • Mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • the individual or subject is a human.
  • An "isolated" antibody is one which has been separated from a component of its natural environment.
  • an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g., SDS- PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC).
  • electrophoretic e.g., SDS- PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatographic e.g., ion exchange or reverse phase HPLC
  • nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment.
  • An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
  • isolated nucleic acid encoding an anti-HERl antibody refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
  • monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts.
  • polyclonal antibody preparations typically include different antibodies directed against different determinants (epitopes)
  • each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including but not limited to the hybridoma method, recombinant DNA methods, phage-display methods, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary methods for making monoclonal antibodies being described herein.
  • the term “Mab” refers to monoclonal antibodies, whereas the term “hMab” refers to humanized variants of such monoclonal antibodies.
  • a “naked antibody” refers to an antibody that is not conjugated to a heterologous moiety (e.g., a cytotoxic moiety) or radiolabel.
  • the naked antibody may be present in a pharmaceutical formulation.(Include if Prior art has immunoconjugates).
  • “Native antibodies” refer to naturally occurring immunoglobulin molecules with varying structures. For example, native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CHI, CH2, and
  • each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain.
  • VL variable region
  • CL constant light
  • the light chain of an antibody may be assigned to one of two types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequence of its constant domain.
  • Percent (%) amino acid sequence identity with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity.
  • Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For purposes herein, however, % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2.
  • the ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No.
  • the ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code.
  • the ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
  • % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows:
  • pharmaceutical formulation refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • HER1 refers to any native HER1 from any vertebrate source, including mammals such as primates (e.g. humans) and rodents (e.g., mice and rats), unless otherwise indicated.
  • the term encompasses "full-length,” unprocessed HER1 as well as any form of HER1 that results from processing in the cell.
  • the term also encompasses naturally occurring variants of HER1, e.g., splice variants or allelic variants.
  • the amino acid sequence of an exemplary human HER1 is shown in SEQ ID NO:2.
  • Human HER1 (also known as epidermal growth factor receptor EGFR or Erb-Bl,) is a 170 kDa transmembrane receptor encoded by the c-erbB proto-oncogene, and exhibits intrinsic tyrosine kinase activity (Modjtahedi, H., et al, Br. J. Cancer 73 (1996) 228-235; Herbst, R.S., and Shin, D.M., Cancer 94 (2002) 1593-1611).
  • SwissProt database entry P00533 provides the sequence of FIERI (SEQ ID NO: 2).
  • FIERI FIERI
  • HER1 is known to bind ligands including a), epidermal growth factor (EGF) (SEQ ID NO: 4), transforming growth factor-a (TGF), amphiregulin, heparin-binding EGF (hb-EGF), betacellulin, and epiregulin (Herbst, R.S., and Shin, D.M., Cancer 94 (2002) 1593-1611;
  • HER1 regulates numerous cellular processes via tyrosine-kinase mediated signal transduction pathways, including, but not limited to, activation of signal transduction pathways that control cell proliferation, differentiation, cell survival, apoptosis, angiogenesis, mitogenesis, and metastasis (Atalay, G., et al, Ann. Oncology 14 (2003) 1346-
  • Epidermal growth factor refers to human epidermal growth factor (EGF, hEGF) (SEQ ID NO: 4).
  • treatment refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • antibodies of the invention are used to delay development of a disease or to slow the progression of a disease.
  • variable region refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen.
  • the variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs).
  • FRs conserved framework regions
  • HVRs hypervariable regions
  • antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Portolano, S. et al, J. Immunol. 150 (1993) 880-887; Clackson, T. et al, Nature 352 (1991) 624-628).
  • vector refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked.
  • the term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced.
  • Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors".
  • the invention is based, in part, on the finding that using the beta- hairpins of HERl functionally presented in a 3-dimensional orientation within SlyD scaffolds (see e.g Figure 2, and the polypeptides of SEQ ID NOs. 12 to 16, and 18) it was possible to select antibodies which are specific for the beta-hairpin of HERl (and do not crossreact of HER2, HER3 and/or HER4).
  • the invention provides an antibody that binds to human HERl , wherein the antibody binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NO: l) of human HERl .
  • Antibodies of the invention are useful, e.g., for the diagnosis or treatment of cancer.
  • the invention provides an isolated antigen binding protein that binds to human HERl , wherein the antigen binding protein binds to a polypeptide selected from the group consisting of:
  • the invention further provides an isolated antigen binding protein that binds to human HERl , wherein the antigen binding protein binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NO: l) which is comprised in a polypeptide selected from the group consisting of:
  • the invention further provides an isolated antigen binding protein that binds to human HER1 , a) wherein the antigen binding protein binds to a polypeptide of
  • the invention further provides an isolated antigen binding protein that binds to human HER1 , wherein the antigen binding protein binds to a polypeptide of
  • SEQ ID NO: 1 TtSlyDcas in an ELISA assay, wherein TtSlyDcys-HERl and TtSlyDcas were immobilized at a concentration of 0.5 ⁇ g/ml.
  • the ELISA signal was detected with a Horse radish peroxidase (HRP)- labeled F(ab " ) 2 goat anti-mouse Fey and 2,2'-Azino-di-[3-ethylbenzthiazoline sulfonate (6)] diammonium salt (ABTS) was used as a HRP-substrate.
  • HRP Horse radish peroxidase
  • the invention further provides an isolated antigen binding protein that binds to human HER1 , a) wherein the antigen binding protein binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NO: l) which is comprised in a polypeptide of SEQ ID NO: 13 (TtSlyDcys-Herl).
  • the invention provides an isolated antibody that binds to human HER1 , a) wherein the antibody binds to a polypeptide selected from the group consisting of:
  • the invention further provides an isolated antibody that binds to human HERl , a) wherein the antibody binds within an amino acid sequence of
  • PPLMLYNPTTYQMDVNPEGK (SEQ ID NO: l) which is comprised in a polypeptide selected from the group consisting of:
  • the invention further provides an isolated antibody that binds to human HERl , a) wherein the antibody binds to a polypeptide of
  • the invention further provides an isolated antibody that binds to human HERl , a) wherein the antibody binds within an amino acid sequence of
  • PPLMLYNPTTYQMDVNPEGK (SEQ ID NO: l) which is comprised in a polypeptide of SEQ ID NO: 13 (TtSlyDcys-Herl).
  • the invention provides an isolated antibody that binds to human HERl , wherein the antibody binds within an amino acid sequence of
  • the invention provides an isolated antibody that binds to human HERl , wherein the antibody has one or more of the following properties (also each combination of each single property is contemplated herein): a) the antibody binds to the amino acid sequence of SEQ ID NO: 1 ; and/or b) the antibody binds to the amino acid sequence SEQ ID NO: l in activated HERl ; and/or c) the antibody binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NO: l) which is comprised in a polypeptide selected from the group consisting of:
  • the antibody binds to a polypeptide with a length of 15 amino acids comprising the amino acid sequence TYQMDVNPEG (SEQ ID NO: 19); and /or h) binds to a polypeptide consisting of TYQMDVNPEG (SEQ ID NO: 19);and/or i) the antibody binds to a polypeptide with a length of 15 amino acids comprising the amino acid sequence MLYNPTTYQ (SEQ ID NO:20); and /or j) binds to a polypeptide consisting of MLYNPTTYQ (SEQ ID NO:20); and/or k) does not induce phosphorylation of HERl in A549 cancer cells in
  • the invention provides an isolated antibody that binds to human HERl, wherein the antibody the antibody binds to a polypeptide with a length of 15 amino acids comprising the amino acid sequence MLYNPTTYQ (SEQ ID NO:20).
  • the invention provides an isolated antibody that binds to human HERl, wherein the antibody the antibody binds to a polypeptide with a length of 15 amino acids comprising the amino acid sequence TYQMDVNPEG (SEQ ID NO: 19).
  • the invention provides an anti-HERl antibody comprising all six HVRs selected from the group consisting of: i) deposited antibody MAK ⁇ HER1-DIB> M-50.097.14 (DSM
  • HVRs are determined according to Kabat.
  • the invention provides an anti-HERl antibody comprising all six HVRs selected from the group consisting of: i) deposited antibody MAK ⁇ HER1-DIB> M-50.097.14 (DSM
  • HVRs are determined according to Kabat.
  • the invention provides an anti-HERl antibody comprising all six HVRs selected from the group consisting of: i) deposited antibody MAK ⁇ HER1-DIB> M-50.097.14 (DSM
  • the antibody has one or more of the following properties: a) the antibody binds to the amino acid sequence of SEQ ID NO: 1 ; and/or b) the antibody binds to the amino acid sequence SEQ ID NO: l in activated HER1 ; and/or c) the antibody binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NO: l) which is comprised in a polypeptide selected from the group consisting of:
  • the antibody binds to a polypeptide with a length of 15 amino acids comprising the amino acid sequence TYQMDVNPEG (SEQ ID NO: 19); and /or h) binds to a polypeptide consisting of TYQMDVNPEG (SEQ ID NO: 19); and/or i) the antibody binds to a polypeptide with a length of 15 amino acids comprising the amino acid sequence MLYNPTTYQ (SEQ ID NO:20); and /or j) binds to a polypeptide consisting of MLYNPTTYQ (SEQ ID NO:20); and/or k) does not induce phosphorylation of HERl in A549 cancer cells in
  • HVRs are determined according to Kabat.
  • the invention provides an antibody that binds to the same epitope as an anti-HERl antibody provided herein.
  • an antibody is provided that binds to the same epitope as an anti- HERl antibody selected from the group consisting of : i) deposited antibody MAK ⁇ HER1-DIB> M-50.097.14 (DSM
  • an antibody that binds to the same as anti- HERl antibody: i) deposited antibody MAK ⁇ HER1-DIB> M-50.097.14 (DSM
  • the invention provides an antibody that competes for binding to human HERl with an anti-HERl antibody provided herein.
  • an antibody is provided that competes for binding to human HERl with an anti- HERl antibody selected from the group consisting of : deposited antibody MAK ⁇ HER1-DIB> M-50.097.14 (DSM ACC3240);
  • an antibody that competes for binding to human HERl with anti- HERl antibody: i) deposited antibody MAK ⁇ HER1-DIB> M-50.097.14 (DSM
  • an antibody that binds to an epitope within a fragment of human HERl consisting of amino acids MLYNPTTYQ (SEQ ID NO:20). In certain embodiments, an antibody is provided that binds to an epitope within a fragment of human HER1 consisting of amino acids TYQMDVNPEG (SEQ ID NO: 19).
  • the antibody is of IgGl or IgG4 isotype.
  • the antibody comprises constant domains of human origin (human constant domains.). Typical human constant regions within the meaning of the present invention comprising the respective human constant domains have the amino acid sequences of SEQ ID NO: 21 to SEQ ID NO:26 (which partly comprise amino acid substitutions). 1. Antibody Affinity
  • an antibody provided herein has a dissociation constant KD of ⁇ 1 ⁇ , ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g. 10 "8 M or less, e.g. from 10 "8 M to 10 "13 M, e.g., from 10 "9 M to 10 "13 M).
  • KD is measured using surface plasmon resonance assays using a BIACORE ® ) at 25°C with immobilized antigen CM5 chips at ⁇ 10 response units (RU). Briefly, carboxymethylated dextran biosensor chips (CM5, BIACORE, Inc.) are activated with N-ethyl-N'- (3-dimethylaminopropyl)- carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions.
  • CM5 carboxymethylated dextran biosensor chips
  • EDC N-ethyl-N'- (3-dimethylaminopropyl)- carbodiimide hydrochloride
  • NHS N-hydroxysuccinimide
  • Antigen is diluted with 10 mM sodium acetate, pH 4.8, to 5 ⁇ g/ml (-0.2 ⁇ ) before injection at a flow rate of 5 ⁇ /minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine is injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of Fab (0.78 nM to 500 nM) are injected in PBS with 0.05% polysorbate 20 (TWEEN-20TM) surfactant (PBST) at 25°C at a flow rate of approximately 25 ⁇ /min.
  • TWEEN-20TM polysorbate 20
  • association rates (k on or ka) and dissociation rates (k 0 ff or kd) are calculated using a simple one-to-one Langmuir binding model (BIACORE Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgrams.
  • the equilibrium dissociation constant KD is calculated as the ratio kd/ka ( k 0 f k on ) See, e.g., Chen, Y. et al, J. Mol. Biol. 293 (1999) 865-881.
  • an antibody provided herein is an antibody fragment.
  • Antibody fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab') 2 , Fv, and scFv fragments, and other fragments described below.
  • Fab fragment-specific antibody fragment
  • Fab' fragment-specific Fab
  • Fab'-SH fragment-specific Fab
  • F(ab') 2 fragment-specific Fab
  • Fv fragment antigen Vpent antibody fragment
  • scFv fragments see, e.g., Plueckthun, A., In; The Pharmacology of Monoclonal Antibodies, Vol. 113, Rosenburg and Moore (eds.), Springer- Verlag, New York (1994), pp.
  • Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 0 404 097; WO 1993/01161; Hudson, P.J. et al, Nat. Med. 9 (2003) 129-134; and Holliger, P. et al, Proc. Natl. Acad. Sci. USA 90 (1993) 6444-6448. Triabodies and tetrabodies are also described in Hudson, P.J. et al, Nat. Med. 9 (20039 129-134).
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody.
  • a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., U.S. Patent No. 6,248,516 Bl).
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
  • recombinant host cells e.g. E. coli or phage
  • an antibody provided herein is a chimeric antibody.
  • chimeric antibodies are described, e.g., in U.S. Patent No. 4,816,567; and Morrison, S.L. et al., Proc. Natl. Acad. Sci. USA 81 (1984) 6851-6855).
  • a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region.
  • a chimeric antibody is a "class switched" antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen- binding fragments thereof.
  • a chimeric antibody is a humanized antibody.
  • a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody.
  • a humanized antibody comprises one or more variable domains in which
  • HVRs e.g., CDRs, (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof) are derived from human antibody sequences.
  • a humanized antibody optionally will also comprise at least a portion of a human constant region.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the HVR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • an antibody provided herein is a human antibody.
  • Human antibodies can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk, M.A. and van de Winkel, J.G., Curr. Opin. Pharmacol. 5 (2001) 368-374 and Lonberg, N., Curr. Opin. Immunol. 20 (2008) 450-459.
  • Human antibodies may be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge.
  • Such animals typically contain all or a portion of the human immunoglobulin loci, which replace the endogenous immunoglobulin loci, or which are present extrachromosomally or integrated randomly into the animal's chromosomes. In such transgenic mice, the endogenous immunoglobulin loci have generally been inactivated.
  • Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeloma cell lines for the production of human monoclonal antibodies have been described. (See, e.g., Kozbor, D., J. Immunol.
  • Human antibodies may also be generated by isolating Fv clone variable domain sequences selected from human-derived phage display libraries. Such variable domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described below.
  • Antibodies of the invention may be isolated by screening combinatorial libraries for antibodies with the desired activity or activities. For example, a variety of methods are known in the art for generating phage display libraries and screening such libraries for antibodies possessing the desired binding characteristics. Such methods are reviewed, e.g., in Hoogenboom, H.R. et al, Methods in Molecular Biology 178 (2001) 1-37 and further described, e.g., in the McCafferty, J. et al, Nature 348 (1990) 552-554; Clackson, T. et al, Nature 352 (1991) 624-628;
  • phage display methods repertoires of VH and VL genes are separately cloned by polymerase chain reaction (PCR) and recombined randomly in phage libraries, which can then be screened for antigen-binding phage as described in Winter, G. et al., Ann. Rev. Immunol. 12 (1994) 433-455.
  • Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments.
  • Libraries from immunized sources provide high-affinity antibodies to the immunogen without the requirement of constructing hybridomas.
  • the naive repertoire can be cloned (e.g., from human) to provide a single source of antibodies to a wide range of non-self and also self antigens without any immunization as described by Griffiths, A.D. et al, EMBO J. 12 (1993) 725-734.
  • naive libraries can also be made synthetically by cloning non-rearranged V-gene segments from stem cells, and using PCR primers containing random sequence to encode the highly variable CDR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom, H.R. and Winter, G., J. Mol. Biol. 227 (1992) 381-388.
  • Patent publications describing human antibody phage libraries include, for example: US Patent No. 5,750,373, and US Patent Publication Nos. 2005/0079574, 2005/0119455, 2005/0266000, 2007/0117126, 2007/0160598, 2007/0237764, 2007/0292936, and 2009/0002360.
  • Antibodies or antibody fragments isolated from human antibody libraries are considered human antibodies or human antibody fragments herein.
  • an antibody provided herein is a multispecific antibody, e.g. a bispecific antibody.
  • Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different sites. In certain embodiments, one of the binding specificities is for HERl and the other is for any other antigen.
  • Bispecific antibodies may also be used to localize cytotoxic agents to cells which express HERl . Bispecific antibodies can be prepared as full length antibodies or antibody fragments.
  • Techniques for making multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein, C. and Cuello, A.C., Nature 305 (1983) 537-540, WO 93/08829, and Traunecker, A. et al, EMBO J. 10 (1991) 3655- 3659), and "knob-in-hole” engineering (see, e.g., U.S. Patent No. 5,731,168).
  • Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004); cross- linking two or more antibodies or fragments (see, e.g., US Patent No. 4,676,980, and Brennan, M. et al Science 229 (1985) 81-83); using leucine zippers to produce bi-specific antibodies (see, e.g., Kostelny, S.A. et al, J. Immunol. 148 (1992) 1547-1553; using "diabody” technology for making bispecific antibody fragments (see, e.g., Holliger, P. et al, Proc. Natl. Acad. Sci.
  • the antibody or fragment herein also includes a "Dual Acting Fab” or “DAF” comprising an antigen binding site that binds to HERl as well as another, different antigen (see, US 2008/0069820, for example).
  • the antibody or fragment herein also includes multispecific antibodies described in WO 2009/080251, WO 2009/080252, WO 2009/080253, WO 2009/080254, WO 2010/112193, WO 2010/115589, WO 2010/136172, WO 2010/145792, and WO 2010/145793.
  • amino acid sequence variants of the antibodies provided herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody.
  • Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding. a) Substitution, Insertion, and Deletion Variants
  • antibody variants having one or more amino acid substitutions are provided.
  • Sites of interest for substitutional mutagenesis include the HVRs and FRs.
  • Conservative substitutions are shown in Table 1 under the heading of "preferred substitutions”. More substantial changes are provided in Table 1 under the heading of "exemplary substitutions," and as further described below in reference to amino acid side chain classes.
  • Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased immunogenicity, or improved ADCC or CDC.
  • Amino acids may be grouped according to common side-chain properties:
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. a humanized or human antibody).
  • a parent antibody e.g. a humanized or human antibody
  • the resulting variant(s) selected for further study will have modifications (e.g., improvements) in certain biological properties (e.g., increased affinity, reduced immunogenicity) relative to the parent antibody and/or will have substantially retained certain biological properties of the parent antibody.
  • An exemplary substitutional variant is an affinity matured antibody, which may be conveniently generated, e.g., using phage display-based affinity maturation techniques such as those described herein. Briefly, one or more HVR residues are mutated and the variant antibodies displayed on phage and screened for a particular biological activity (e.g. binding affinity).
  • Alterations may be made in HVRs, e.g., to improve antibody affinity. Such alterations may be made in HVR "hotspots," i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, P.S., Methods Mol. Biol. 207 (2008) 179-196), and/or SDRs (a-CDRs), with the resulting variant VH or VL being tested for binding affinity.
  • HVR "hotspots” i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, P.S., Methods Mol. Biol. 207 (2008) 179-196), and/or SDRs (a-CDRs), with the resulting variant VH or VL being tested for binding affinity.
  • Affinity maturation by constructing and reselecting from secondary libraries has been described, e
  • affinity maturation diversity is introduced into the variable genes chosen for maturation by any of a variety of methods (e.g., error-prone PCR, chain shuffling, or oligonucleotide-directed mutagenesis).
  • a secondary library is then created. The library is then screened to identify any antibody variants with the desired affinity.
  • Another method to introduce diversity involves HVR-directed approaches, in which several HVR residues (e.g., 4-6 residues at a time) are randomized. HVR residues involved in antigen binding may be specifically identified, e.g., using alanine scanning mutagenesis or modeling. CDR-H3 and CDR-L3 in particular are often targeted.
  • substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind antigen.
  • conservative alterations e.g., conservative substitutions as provided herein
  • HVRs Such alterations may be outside of HVR "hotspots" or SDRs.
  • each HVR either is unaltered, or contains no more than one, two or three amino acid substitutions.
  • a useful method for identification of residues or regions of an antibody that may be targeted for mutagenesis is called "alanine scanning mutagenesis" as described by Cunningham, B.C.
  • a residue or group of target residues e.g., charged residues such as arg, asp, his, lys, and glu
  • a neutral or negatively charged amino acid e.g., alanine or polyalanine
  • Further substitutions may be introduced at the amino acid locations demonstrating functional sensitivity to the initial substitutions.
  • a crystal structure of an antigen- antibody complex to identify contact points between the antibody and antigen.
  • Such contact residues and neighboring residues may be targeted or eliminated as candidates for substitution.
  • Variants may be screened to determine whether they contain the desired properties.
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • terminal insertions include an antibody with an N-terminal methionyl residue.
  • Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme (e.g. for ADEPT) or a polypeptide which increases the serum half-life of the antibody.
  • ADEPT enzyme
  • an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated.
  • Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
  • the carbohydrate attached thereto may be altered.
  • Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright, A. and Morrison, S.L., TIBTECH 15 (1997) 26-32.
  • the oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the "stem" of the biantennary oligosaccharide structure.
  • modifications of the oligosaccharide in an antibody of the invention may be made in order to create antibody variants with certain improved properties.
  • antibody variants are provided having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region.
  • the amount of fucose in such antibody may be from 1% to 80%, from 1% to 65%, from
  • the amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e. g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example.
  • Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ⁇ 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies.
  • Such fucosylation variants may have improved ADCC function. See, e.g., US 2003/0157108; US 2004/0093621.
  • Examples of publications related to "defucosylated” or "fucose-deficient" antibody variants include: US 2003/0157108; WO 2000/61739; WO 2001/29246; US 2003/0115614; US 2002/0164328; US 2004/0093621; US 2004/0132140; US 2004/0110704; US 2004/0110282; US 2004/0109865; WO 2003/085119; WO 2003/084570; WO 2005/035586; WO 2005/035778; WO 2005/053742; WO 2002/031140;
  • Examples of cell lines capable of producing defucosylated antibodies include Led 3 CHO cells deficient in protein fucosylation (Ripka, J. et al, Arch. Biochem. Biophys. 249 (1986) 533-545; US 2003/0157108; and WO 2004/056312, especially at Example 11), and knockout cell lines, such as alpha- 1,6-fucosyltransferase gene, FUT8, knockout CHO cells (see, e.g., Yamane- Ohnuki, N. et al, Biotech. Bioeng. 87 (2004) 614-622; Kanda, Y. et al, Biotechnol. Bioeng. 94 (2006) 680-688; and WO 2003/085107).
  • Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc. Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/011878; US Patent No. 6,602,684; and US 2005/0123546. Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided. Such antibody variants may have improved CDC function. Such antibody variants are described, e.g., in WO 1997/30087; WO 1998/58964; and WO 1999/22764. c) Fc region variants
  • one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby generating an Fc region variant.
  • the Fc region variant may comprise a human Fc region sequence (e.g., a human IgGl, IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification (e.g. a substitution) at one or more amino acid positions.
  • the invention contemplates an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half-life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious.
  • In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities.
  • Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FcyR binding (hence likely lacking ADCC activity), but retains FcRn binding ability.
  • NK cells express Fc(RIII only, whereas monocytes express FcgammaRI, FcgammaRII and FcgammaRIII.
  • FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch, J. V. and Kinet, J.P., Annu. Rev. Immunol. 9 (1991) 457-492.
  • Non-limiting examples of in vitro assays to assess ADCC activity of a molecule of interest is described in U.S. Patent No. 5,500,362 (see, e.g. Hellstrom, I. et al, Proc. Natl. Acad. Sci.
  • non-radioactive assays methods may be employed (see, for example, ACTITM non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, CA; and CytoTox 96 ® nonradioactive cytotoxicity assay (Promega, Madison, WI).
  • PBMC peripheral blood mononuclear cells
  • NK Natural Killer
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in an animal model such as that disclosed in Clynes, R. et al, Proc. Natl. Acad. Sci. USA 95 (1998) 652-656.
  • Clq binding assays may also be carried out to confirm that the antibody is unable to bind Clq and hence lacks CDC activity. See, e.g., Clq and C3c binding ELISA in WO 2006/029879 and WO 2005/100402.
  • a CDC assay may be performed (see, for example, Gazzano-Santoro, H. et al., J. Immunol. Methods 202 (1996) 163-171; Cragg, M.S. et al, Blood 101 (2003) 1045-1052; and Cragg, M.S. and M.J. Glennie, Blood 103 (2004) 2738-2743).
  • FcRn binding and in vivo clearance/half-life determinations can also be performed using methods known in the art (see, e.g., Petkova, S.B. et al, Int. Immunol. 18 (2006: 1759-1769).
  • Antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Patent No. 6,737,056).
  • Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called "DANA" Fc mutant with substitution of residues 265 and 297 to alanine
  • an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC, e.g., substitutions at positions 298, 333, and/or 334 of the Fc region (EU numbering of residues).
  • alterations are made in the Fc region that result in altered (i.e., either improved or diminished) Clq binding and/or Complement Dependent Cytotoxicity (CDC), e.g., as described in US Patent No. 6,194,551, WO 99/51642, and Idusogie, E.E. et al, J. Immunol. 164 (2000) 4178-4184.
  • CDC Complement Dependent Cytotoxicity
  • Antibodies with increased half-lives and improved binding to the neonatal Fc receptor (FcRn), which is responsible for the transfer of maternal IgGs to the fetus are described in US 2005/0014934.
  • Those antibodies comprise an Fc region with one or more substitutions therein which improve binding of the Fc region to FcRn.
  • Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US Patent No. 7,371,826).
  • cysteine engineered antibodies e.g., "thioMAbs”
  • one or more residues of an antibody are substituted with cysteine residues.
  • the substituted residues occur at accessible sites of the antibody.
  • reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate, as described further herein.
  • any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; Al 18 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region.
  • Cysteine engineered antibodies may be generated as described, e.g., in U.S. Patent No. 7,521,541.
  • an antibody provided herein may be further modified to contain additional non-proteinaceous moieties that are known in the art and readily available.
  • the moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers.
  • Non-limiting examples of water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3-dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)polyethylene glycol, propropylene glycol homopolymers, prolypropylene oxide/ethylene oxide co- polymers, polyoxyethylated polyols (e.g., glycerol), polyvinyl alcohol, and mixtures thereof.
  • PEG polyethylene glycol
  • copolymers of ethylene glycol/propylene glycol carboxymethylcellulose
  • dextran polyvinyl alcohol
  • Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the number of polymers attached to the antibody may vary, and if more than one polymer is attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
  • conjugates of an antibody and non-proteinaceous moiety that may be selectively heated by exposure to radiation are provided.
  • the non-proteinaceous moiety is a carbon nanotube (Kam, N.W. et al., Proc. Natl. Acad. Sci. USA 102 (2005) 11600-11605).
  • the radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the non-proteinaceous moiety to a temperature at which cells proximal to the antibody-non-proteinaceous moiety are killed.
  • Antibodies may be produced using recombinant methods and compositions, e.g., as described in U.S. Patent No. 4,816,567.
  • isolated nucleic acid encoding an anti-HERl antibody described herein is provided.
  • Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody).
  • one or more vectors e.g., expression vectors
  • a host cell comprising such nucleic acid is provided.
  • a host cell comprises (e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody.
  • the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NS0, Sp20 cell).
  • a method of making an anti-HERl antibody comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).
  • nucleic acid encoding an antibody is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
  • nucleic acid may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
  • Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein.
  • antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed.
  • For expression of antibody fragments and polypeptides in bacteria see, e.g., US 5,648,237, US 5,789,199, and US 5,840,523. (See also Charlton, K.A., In:
  • the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been "humanized,” resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, T.U., Nat. Biotech. 22 (2004) 1409-1414; and Li, H. et al, Nat. Biotech. 24 (2006) 210-215.
  • Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures can also be utilized as hosts. See, e.g., US Patent Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIESTM technology for producing antibodies in transgenic plants).
  • Vertebrate cells may also be used as hosts.
  • mammalian cell lines that are adapted to grow in suspension may be useful.
  • useful mammalian host cell lines are monkey kidney CVl line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham, F.L. et al, J. Gen Virol. 36 (1977) 59-74); baby hamster kidney cells (BHK); mouse Sertoli cells (TM4 cells as described, e.g., in Mather, J.P., Biol. Reprod.
  • monkey kidney cells (CVl); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3 A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather, J.P. et al, Annals N.Y. Acad. Sci. 383 (1982) 44-68; MRC 5 cells; and FS4 cells.
  • Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR " CHO cells (Urlaub, G. et al, Proc. Natl. Acad.
  • Anti-HERl antibodies provided herein may be identified, screened for, or characterized for their physical/chemical properties and/or biological activities by various assays known in the art.
  • One aspect of the invention is a method for selecting an antibody (or antigen binding protein) that binds to human HERl, wherein the antibody(or antigen binding protein) binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NO : 1 ) of human HERl ; wherein a) at least one polypeptide selected from the group consisting of:
  • SEQ ID NO 18 TgSlyDcys-HERl which comprises the amino acid sequence of SEQ ID NO : 1 ; is used to select (in a binding assay) antibodies (or antigen binding proteins), which show binding to the at least one polypeptide under a), and thereby selecting an antigen binding protein, in particular an antibody that binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NO: l) of human HERl .
  • selection methods further comprises a step wherein the selected antibodies are counterscreened with the polypeptides (tested for binding to the polypeptides) selected from the group consisting of:
  • the invention provides an antibody(or antigen binding protein) obtained by such selection method.
  • a method for selecting an antibody(or antigen binding protein) that specifically binds to a human HERl comprising the following steps: a) determining the binding affinity of a plurality of antibodies (or antigen binding proteins) to the B-hairpin of HERl with the amino acid sequence of SEQ ID NO: l , whereby B-hairpin of HERl is presented as polypeptide selected from the group consisting of: i) SEQ ID NO 12 TtSlyDcas-HERl ,
  • SEQ ID NO 18 TgSlyDcys-HERl which comprise the B-hairpin of HERl with the amino acid sequence of SEQ ID NO: l , b) selecting the antibody (or antigen binding protein) having an apparent complex stability above a pre-defined threshold level.
  • an antibody (or antigen binding protein) of the invention is tested for its antigen binding activity, e.g., by known methods such as ELISA, Western blot, including surface plasmon resonance (e.g. BIACORE) , etc.
  • competition assays may be used to identify an antibody that competes with i) deposited antibody MAK ⁇ HER1-DIB> M-50.097.14 (DSM
  • such a competing antibody binds to the same epitope (e.g., a linear or a conformational epitope) that is bound by i) deposited antibody MAK ⁇ HER1-DIB> M-50.097.14 (DSM
  • immobilized HERl is incubated in a solution comprising a first labeled antibody that binds to HERl, respectively (e.g. deposited antibodies MAK ⁇ HER1-DIB> M-50.097.14 (DSM ACC3240MAK ⁇ HER1- DIB>M-50.110.23(DSM ACC3241);MAK ⁇ HER1-DIB> M-37.058.09 (DSM ACC3238);MAK ⁇ HER1-DIB> M-37.186.15 (DSM ACC3239)) and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to HERl .
  • a first labeled antibody that binds to HERl
  • the second antibody may be present in a hybridoma supernatant.
  • immobilized HERl is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody. After incubation under conditions permissive for binding of the first antibody to HERl, excess unbound antibody is removed, and the amount of label associated with immobilized HERl is measured. If the amount of label associated with immobilized HERl is substantially reduced in the test sample relative to the control sample, then that indicates that the second antibody is competing with the first antibody for binding to HERl . See Harlow, E. and Lane, D., Antibodies: A Laboratory Manual, Chapter 14, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (1988).
  • assays are provided for identifying anti-HERl antibodies (or antigen binding proteins) thereof having biological activity.
  • Biological activity may include, e.g., inhibition of HERl phosphorylation, non-agonistic activity with respect to HERl phosphorylation in the absence of EGF, inhibition of cancer cell proliferation of HERl expressing or overexpressing cancer cells, inhibition of HER1/HER1 homodimerization inhibition of HER1/HER2 heterodimerization, (time-dependent) internalization via Western Blot or FACS assay, in vivo tumor growth inhibition in xenograft animal ( e.g. mouse or rat) models with xenografted HERl expressing or overexpressing cancer cells.
  • Antibodies having such biological activity either alone or as immunoconjugates with a cytotoxic agent in vivo and/or in vitro are also provided.
  • an antibody of the invention is tested for such biological activity.
  • Exemplary vitro or in vivo assays for specified biological activities are described in Example 2e, and Examples 5, 6 and 8.
  • the invention also provides immunoconjugates comprising an anti-HERl antibody (or antigen binding protein) described herein conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • cytotoxic agents such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • an immunoconjugate is an antibody-drug conjugate (ADC) in which an antibody is conjugated to one or more drugs, including but not limited to a maytansinoid (see US 5,208,020, US 5,416,064 and EP 0 425 235 Bl); an auristatin such as monomethyl auristatin drug moieties DE and DF (MMAE and MMAF) (see US 5,635,483, US 5,780,588, and US 7,498,298); a dolastatin; a calicheamicin or derivative thereof (see US 5,712,374, US 5,714,586, US 5,739,116, US 5,767,285, US 5,770,701, US 5,770,710, US 5,773,001, and US 5,877,296; Hinman, L.M.
  • ADC antibody-drug conjugate
  • an immunoconjugate comprises an antibody as described herein conjugated to an enzymatically active toxin or fragment thereof, including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
  • an enzymatically active toxin or fragment thereof including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (
  • an immunoconjugate comprises an antibody as described herein conjugated to a Pseudomonas exotoxin A or variants thereof.
  • Pseudomonas exotoxin A or variants thereof are described e.g in WO2011/32022,
  • an immunoconjugate comprises an antibody as described herein conjugated to a radioactive atom to form a radioconjugate.
  • a variety of radioactive isotopes are available for the production of radioconjugates. Examples include At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu.
  • the radioconjugate When used for detection, it may comprise a radioactive atom for scintigraphic studies, for example TC 99m or I 123 , or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, MRI), such as iodine- 123 again, iodine-131, indium-I l l, fluorine- 19, carbon- 13, nitrogen- 15, oxygen- 17, gadolinium, manganese or iron.
  • NMR nuclear magnetic resonance
  • Conjugates of an antibody and cytotoxic agent may be made a) either using recombination expression techniques (e.g for the expression of amino acid sequence based toxines fused to a Fab or Fv antibody fragment e.g. in E.coli) or b) using polypeptide coupling techniques (like sortase enzyme based coupling of amino acid sequence based toxines to a Fab or Fv antibody fragment) or c) using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2- pyridyldithio) propionate (SPDP), succinimidyl-4-(N-maleimidomethyl) cyclohexane-l-carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HC1), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde
  • a ricin immunotoxin can be prepared as described in Vitetta, E.S. et al, Science 238 (1987) 1098-1104.
  • Carbon- 14-labeled 1- isothiocyanatobenzyl-3-methyldiethylene triamine pentaacetic acid is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See WO 94/11026.
  • the linker may be a "cleavable linker" facilitating release of a cytotoxic drug in the cell.
  • an acid-labile linker, peptidase-sensitive linker, photolabile linker, dimethyl linker or disulfide-containing linker (Chari, R.V.
  • the immunuoconjugates or ADCs herein expressly contemplate, but are not limited to such conjugates prepared with cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo-SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB (succinimidyl-(4- vinylsulfone)benzoate) which are commercially available (e.g., from Pierce
  • any of the anti-HERl antibodies (or antigen binding proteins) provided herein is useful for detecting the presence of HERl, respectively in a biological sample.
  • the term "detecting” as used herein encompasses quantitative or qualitative detection.
  • a biological sample comprises a cell or tissue, such as tumor tissues.
  • an anti-HERl antibody for use in a method of diagnosis or detection is provided.
  • a method of detecting the presence of HERl, respectively, in a biological sample comprises contacting the biological sample with an anti-HERl antibody as described herein under conditions permissive for binding of the anti-HERl antibody to HERl, respectively, and detecting whether a complex is formed between the anti-HERl antibody and HERl, respectively.
  • Such method may be an in vitro or in vivo method.
  • an anti-HERl antibody is used to select subjects eligible for therapy with an the anti-HERl antibodies antibody, e.g. where HERl, respectively are both biomarkers for selection of patients.
  • Exemplary disorders that may be diagnosed using an antibody of the invention include cancer.
  • labeled anti-HERl antibodies include, but are not limited to, labels or moieties that are detected directly (such as fluorescent, chromophoric, electron-dense, chemiluminescent, and radioactive labels), as well as moieties, such as enzymes or ligands, that are detected indirectly, e.g., through an enzymatic reaction or molecular interaction.
  • labels or moieties that are detected directly such as fluorescent, chromophoric, electron-dense, chemiluminescent, and radioactive labels
  • moieties such as enzymes or ligands
  • 32 14 125 3 131 include, but are not limited to, the radioisotopes P, C, I, H, and I, fluorophores such as rare earth chelates or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, luceriferases, e.g., firefly luciferase and bacterial luciferase (U.S. Patent No.
  • compositions of an anti-HERl antibody (or antigen binding protein) as described herein are prepared by mixing such antibody having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Remington's Pharmaceutical Sciences, 16th edition, Osol, A. (ed.) (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyl dimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone); amino acids such as glycine, glutamine, asparagine, histidine, argin
  • sHASEGP soluble neutral-active hyaluronidase glycoproteins
  • rhuPH20 HYLENEX®, Baxter International, Inc.
  • Certain exemplary sHASEGPs and methods of use, including rhuPH20, are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968.
  • a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
  • Exemplary lyophilized antibody formulations are described in US Patent No. 6,267,958.
  • Aqueous antibody formulations include those described in US Patent No. 6,171 ,586 and WO 2006/044908, the latter formulations including a histidine-acetate buffer.
  • the formulation herein may also contain more than one active ingredients as necessary for the particular indication being treated.
  • Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methyl methacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • macroemulsions for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semi-permeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • the formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes. G. Therapeutic Methods and Compositions
  • any of the anti-HERl antibodies (or antigen binding proteins) or immunoconjugates of the anti-HERl antibodies (or antigen binding protein) conjugated to a cytotoxic agent, provided herein may be used in therapeutic methods.
  • an anti-HERl antibody or immunoconjugate of the anti-HERl antibody conjugated to a cytotoxic agent for use as a medicament is provided.
  • an anti-HERl antibody or immunoconjugate of the anti-HERl antibody conjugated to a cytotoxic agent for use in treating cancer is provided.
  • an anti-HERl antibody or immunoconjugates of the anti- HER1 antibody conjugated to a cytotoxic agent for use in a method of treatment is provided.
  • the invention provides an anti-HERl antibody or immunoconjugate of the anti-HERl antibody conjugated to a cytotoxic agent for use in a method of treating an individual having cancer comprising administering to the individual an effective amount of the anti-HERl antibody or the immunoconjugate of the anti-HERl antibody conjugated to a cytotoxic agent.
  • the invention provides an anti-HERl antibody or immunoconjugate of the anti-HERl antibody conjugated to a cytotoxic agent for use in inducing apoptosis in a cancer cell/ or inhibiting cancer cell proliferation.
  • the invention provides an anti-HERl antibody or immunoconjugate of the anti-HERl antibody conjugated to a cytotoxic agent for use in a method of inducing apoptosis in a cancer cell/ or inhibiting cancer cell proliferation in an individual comprising administering to the individual an effective of the anti-HERl antibody or immunoconjugate of the anti-HERl antibodies conjugated to a cytotoxic agent to induce apoptosis in a cancer cell/ or to inhibit cancer cell proliferation.
  • An "individual" according to any of the above embodiments is preferably a human.
  • the invention provides for the use of an anti-HERl antibody or an immunoconjugate of the anti-HERl antibody conjugated to a cytotoxic agent in the manufacture or preparation of a medicament.
  • the medicament is for treatment of cancer.
  • the medicament is for use in a method of treating cancer comprising administering to an individual having cancer an effective amount of the medicament.
  • the medicament is for inducing apoptosis in a cancer cell/ or inhibiting cancer cell proliferation.
  • the medicament is for use in a method of inducing apoptosis in a cancer cell/ or inhibiting cancer cell proliferation in an individual suffering from cancer comprising administering to the individual an amount effective of the medicament to induce apoptosis in a cancer cell/ or to inhibit cancer cell proliferation.
  • An "individual" according to any of the above embodiments may be a human.
  • the invention provides a method for treating cancer.
  • the method comprises administering to an individual having cancer an effective amount of an anti-HERl antibody.
  • An "individual" according to any of the above embodiments may be a human.
  • the invention provides a method for inducing apoptosis in a cancer cell/ or inhibiting cancer cell proliferation in an individual suffering from cancer.
  • the method comprises administering to the individual an effective amount of an anti-HERl antibody or an immunoconjugate of the anti- HERl antibody conjugated to a cytotoxic compound to induce apoptosis in a cancer cell/ or to inhibit cancer cell proliferation in the individual suffering from cancer.
  • an "individual" is a human.
  • the invention provides pharmaceutical formulations comprising any of the anti-HERl antibodies provided herein, e.g., for use in any of the above therapeutic methods.
  • a pharmaceutical formulation comprises any of the anti-HERl antibodies provided herein and a pharmaceutically acceptable carrier.
  • An antibody of the invention (and any additional therapeutic agent) can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic. Various dosing schedules including but not limited to single or multiple administrations over various time- points, bolus administration, and pulse infusion are contemplated herein.
  • Antibodies of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above.
  • an antibody of the invention when used alone or in combination with one or more other additional therapeutic agents, will depend on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the antibody is suitably administered to the patient at one time or over a series of treatments.
  • about 1 ⁇ g/kg to 15 mg/kg (e.g. 0.5mg/kg - 10 mg/kg) of antibody can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • One typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment would generally be sustained until a desired suppression of disease symptoms occurs.
  • One exemplary dosage of the antibody would be in the range from about 0.05 mg/kg to about 10 mg/kg.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the antibody).
  • An initial higher loading dose, followed by one or more lower doses may be administered.
  • An exemplary dosing regimen comprises administering an initial loading dose of about 4 mg/kg, followed by a weekly maintenance dose of about 2 mg/kg of the antibody.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided.
  • the article of manufacture comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is an antibody of the invention.
  • the label or package insert indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an antibody of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically- acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate- buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • a pharmaceutically- acceptable buffer such as bacteriostatic water for injection (BWFI), phosphate- buffered saline, Ringer's solution and dextrose solution.
  • any of the above articles of manufacture may include an immunoconjugate of the invention in place of or in addition to an anti-HERl antibody.
  • SEQ ID NO: 21 human kappa light chain constant region
  • SEQ ID NO: 22 human lambda light chain constant region
  • SEQ ID NO: 23 human heavy chain constant region derived from IgGl
  • SEQ ID NO: 24 human heavy chain constant region derived from IgGl mutated on L234A and L235A
  • SEQ ID NO: 25 human heavy chain constant region derived from IgGl mutated on L234A, L235A and P329G
  • SEQ ID NO: 26 human heavy chain constant region derived from IgG4
  • SEQ ID NO 18 TgSlyDcys-HERl which comprises the amino acid sequence of SEQ ID NO:l; is used to select antigen binding proteins, which show binding to the at least one polypeptide under a) and thereby selecting an antigen binding protein that binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NOT) of human HER1.
  • An antigen binding protein obtained by the selection method of embodiment 1.
  • An isolated antigen binding protein that binds to human HERl wherein the antigen binding protein binds within an amino acid sequence of PPLMLYNPTTYQMDVNPEGK (SEQ ID NO: l) which is comprised in a polypeptide of SEQ ID NO: 13 (TtSlyDcys-Herl).
  • antigen binding protein of embodiments 4 or 5 wherein the antigen binding protein is an antibody is an antibody.
  • an isolated antibody of any one of the preceding embodiments wherein the antibody shows more than 70 percent internalization of HERl in the presence of EGF after 2 h after incubation with the antibody in a Western Blot assay with HERl expressing A549 cells (ATCC CCL-185) and shows less than 55 percent internalization of HERl in the absence of EGF after 2 h after incubation with the antibody in a Western Blot assay with HERl expressing A549 cells.
  • An isolated antibody that binds to human HERl wherein the antibody binds to a polypeptide with a length of 15 amino acids, the polypeptide comprising the amino acid sequence of TYQMDVNPEG (SEQ ID NO: 19).
  • An isolated antibody that binds to human HERl wherein the antibody binds to a polypeptide with a length of 15 amino acids, the polypeptide comprising the amino acid sequence of MLYNPTTYQ (SEQ ID NO:20).
  • the antibody of embodiments 6 to 10 which is a human, humanized, or chimeric antibody.
  • the antibody of embodiments 6 to 10 which is an antibody fragment that binds human HERl .
  • an isolated antibody that binds to human HERl wherein the antibody comprises i) (a) HVR-H 1 ; (b) HVR-H2; (c) HVR-H3 ;(d) HVR-L 1 ; (e) HVR-L2; and (f) HVR-L3 of deposited antibody MAK ⁇ HER1-DIB> M-
  • An immunoconjugate comprising the antibody of any one of embodiments 6 to 13 and a cytotoxic agent.
  • a pharmaceutical formulation comprising the antibody of any one of embodiments 6 to 13, or the immunoconjugate of embodiment 16, and a pharmaceutically acceptable carrier.
  • immunoconjugate of embodiment 16 in the manufacture of a medicament.
  • HER1/HER2 and/or HER 1 /HER 1 dimerization are HER1/HER2 and/or HER 1 /HER 1 dimerization.
  • a method of treating an individual having cancer comprising administering to the individual an effective amount of the antibody of any one of the preceding embodiments, or an immunoconjugate comprising the antibody of any one of the preceding embodiments and a cytotoxic agent.
  • a method of inducing apoptosis in a cancer cell in an individual suffering from cancer comprising administering to the individual an effective amount of an immunoconjugate comprising the antibody of any one of the preceding embodiments and a cytotoxic agent, thereby inducing apoptosis in a cancer cell in the individual.
  • Isolated nucleic acid encoding the antibody of any one of embodiments 6 to 11. 27.
  • a host cell comprising the nucleic acid of embodiment 26.
  • a method of producing an antibody comprising culturing the host cell of embodiment 27 so that the antibody is produced.
  • polypeptide comprises the amino acid sequence of SEQ ID NO: 1.
  • Desired gene segments were prepared from oligonucleotides made by chemical synthesis.
  • the 400 - 1600 bp long gene segments, which were flanked by singular restriction endonuclease cleavage sites, were assembled by annealing and ligating oligonucleotides including PCR amplification and subsequently cloned via the indicated restriction sites e.g. EcoRI/ Blpl or Bsml/Xhol into the expression vectors described below.
  • the DNA sequences of the subcloned gene fragments were confirmed by DNA sequencing. Gene synthesis fragments were ordered according to given specifications at Geneart (Regensburg, Germany). DNA sequence determination
  • DNA sequences were determined by double strand sequencing performed at Sequiserve GmbH (Vaterstetten, Germany).
  • Infomax's Vector NT1 Advance suite version 11.5.0 was used for sequence creation, mapping, analysis, annotation and illustration.
  • HER1 B-HairpinHERl was grafted into a SlyD polypeptide framework comprising a FKBP domain.
  • the grafted B-Hairpins are freely accessible in contrast to the hidden structure in the native unactivated conformation of HER1 (in the absence of ligand as e.g. EGF) (see Figure 1 and 2, where the B-Hairpin of HER1 is hidden).
  • E. coli BL21 (DE3) cells transformed with the particular expression plasmid were grown at 37°C in LB medium containing the respective antibiotic for selective growth (Kanamycin 30 ⁇ g/ml, or Ampicillin (100 ⁇ g/ml)) to an OD600 of 1.5, and cytosolic overexpression was induced by adding 1 mM isopropyl-B-D- thiogalactoside (IPTG). Three hours after induction, cells were harvested by centrifugation (20 min at 5,000 g), frozen and stored at -20 °C.
  • the frozen pellet was resuspended in chilled 50 mM sodium phosphate buffer (pH 8.0) supplemented with 7 M GdmCl and 5 mM imidazole. Thereafter the suspension was stirred for 2-10 hours on ice to complete cell lysis. After centrifugation (25,000 g, 1 h) and filtration (cellulose nitrate membrane, 8.0 ⁇ , 1.2 ⁇ , 0.2 ⁇ ), the lysate was applied onto a Ni-NTA column equilibrated with the lysis buffer.
  • the imidazole concentration was raised to 10 mM (in 50 mM sodium phosphate buffer (pH 8.0) comprising 7 M GdmCl) and 5 mM TCEP was added in order to keep the thiol moieties in a reduced form and to prevent premature disulfide bridging. At least 15 to 20 volumes of the reducing washing buffer were applied. Thereafter, the GdmCl solution was replaced by 50 mM sodium phosphate buffer (pH 8.0) comprising 100 mM NaCl, lO mM imidazole, and 5 mM TCEP to induce conformational refolding of the matrix-bound SlyD fusion polypeptide.
  • a protease inhibitor cocktail (Complete ® EDTA-free, Roche) was added to the refolding buffer. A total of 15 to 20 column volumes of refolding buffer were applied in an overnight procedure. Thereafter, both TCEP and the Complete ® EDTA-free inhibitor cocktail were removed by washing with 10 column volumes 50 mM sodium phosphate buffer (pH 8.0) comprising 100 mM NaCl and 10 mM imidazole. In the last washing step, the imidazole concentration was raised to 30 mM (10 column volumes) in order to remove tenacious contaminants. The refolded polypeptide was then eluted by applying 250 mM imidazole in the same buffer.
  • Protein-containing fractions were assessed for purity by Tricine-SDS-PAGE (Schaegger, H. and von Jagow, G., Anal. Biochem. 166 (1987) 368-379). Subsequently, the protein was subjected to size-exclusion-chromatography (SuperdexTM HiLoad, Amersham Pharmacia) using potassium phosphate as the buffer system (50 mM potassium phosphate buffer (pH 7.0), 100 mM KC1, 0.5 mM EDTA). Finally, the protein-containing fractions were pooled and concentrated in an Amicon cell (YM10) to a concentration of ⁇ 5 mg/ml. Exemplarily SDS-PAGE analysis of Ni-NTA purification of TtSlyDcas-HERl is shown in Figure 3 and SEC elution profile of a Ni-NTA purified fraction of Thermus thermophilus SlyDcas-
  • HER1 is shown in Figure 4.
  • the TtSlyDcas-HERl fusion polypeptide could be purified successfully as a soluble and stable polypeptide in its monomeric form.
  • the final yield was quantified at 30 mg purified protein from fraction 11 and 12.
  • Table 2 Summary of the amino acid sequences of the developed SlyD-based epitope scaffolds (which carry the HERl dimerization domain fragment HERl as insert.
  • TtSlyDcas and TgSlyDAIF were used as negative controls in the ELISA screening (without the HERl dimerization domain fragment ( ⁇ -Hairpin of HERl as insert).
  • TtSlyDcas Nt- SEQ ID MRSKVGQDKVVTIRYTLQVEGEVLDQGELSYLHGHRNLIPGL NO: 1 1 EEALEGREEGEAFQAHVPAEKAYGAGSGSSGKDLDFQVEVV
  • mice For the generation of antibodies against the B-hairpin of HERl , Balb/C, NMRI or SJL mice were immunized with different antigens.
  • antigens the following proteins were used: full length HERl ECD, or the epitope scaffold proteins TtSlyDcas-HERl , TtSlyDcys-HERl and TtSlyD(GSG)-HERl .
  • the TtSlyDcas- HER1 variant represents the first generation epitope scaffold, used for generation of HERl dimerization domain specific antibodies.
  • mice All mice were subjected to 3 immunizations at the time points 0, 6 and 10 weeks after start of the immunization campaign. At each time point each mouse was immunized with 100 ⁇ g endotoxin free immunogen dissolved in 100 ⁇ PBS. For the first immunization the immunogen was mixed with 100 ⁇ CFA. For the second and third immunization the immunogen was mixed with IFA. The first and the third immunization were applied via the intraperitoneal route, the second immunization was applied subcutaneously. 2 and 3 days prior to the preparation of spleenocyte for antibody development using hybridoma technology, the mice were subjected to intravenous booster immunizations with 12.5 ⁇ g immunogen in 100 ⁇ PBS and without adjuvant. Titer analysis
  • ECD was immobilized at a concentration of 1 ⁇ g/ml and the scaffold proteins TtSlyDcas-HERl , TtSlyDcys-HERl , TtSlyD(GSG)-HERl , TtSlyDcas, TgSlyDAIF and TgSlyDcys-HERl were used at a concentration of 0.5 ⁇ g/ml.
  • the scaffold proteins TtSlyDcas and TgSlyDAIF were used as negative controls.
  • the sera from each mouse were diluted in PBS with 1% BSA and the dilutions were added to the plates.
  • the sera were tested at dilutions 1 :300, 1 :900, 1 :2700, 1 :8100, 1 :24300, 1 :72900, 1 :218700 and 1 :656100.
  • Bound antibody was detected with a HRP- labeled F(ab " ) 2 goat anti-mouse Fey (Dianova) and ABTS (Roche) as a substrate.
  • mice immunized with trastoxin developed antibodies against the HERl ⁇ -hairpin domain.
  • mice immunized with trastoxin developed antibodies against the HERl ⁇ -hairpin domain.
  • HERl ECD this can be shown by titration against one of the scaffold proteins containing the dimerization ⁇ -hairpin loop.
  • the strongly reduced signal can be explained by the fact, that the majority of antibodies raised by immunization with HERl ECD are targeting other parts within the ECD and only a small fraction is binding to the dimerization ⁇ -hairpin domain.
  • the fraction of antibodies targeting the loop can be shown by titration against Herl ECD (positive control) and titration against an control scaffold without HERl insertion (negative control).
  • the use of the here described epitope scaffold technology offers in principal two strategies for the development of antibodies targeting the HERl dimerization domain ( ⁇ -Hairpins of HERl).
  • One strategy is to immunize with the full length HERl ECD and to use the scaffolds to screen for the dimerization domain specific antibodies.
  • the other strategy is the direct use of the scaffold for immunization and to use the HERl ECD, a scaffold with another backbone or a scaffold without insertion for counter screening.
  • Antibodies were developed with hybridoma technology by fusing primary B-cells with P3X63Ag8.653 myeloma cells.
  • mice 2 days after the final booster immunization, immunized mice were sacrificed and spleen cell populations were prepared.
  • the spleenocytes were fused with P3X63Ag8.653 by using the PEG fusion technology.
  • the cellular batch culture from the fusion was incubated overnight at 37°C under 5% C0 2 .
  • the following day the cellular batch containing fused cells was centrifuged for 10 min at 300 g. Thereafter, the cells were suspended in hybridoma selection media supplemented with O. lx azaserine- hypoxanthine (Sigma) and were seeded at a concentration of 2.5xl0 4 cells per well in 96well plates.
  • the plates were cultured for at least 1 week at 37°C under 5% C0 2 . 3 days prior to ELISA analysis the selection media was changed.
  • HERl ECD was immobilized at a concentration of 1 ⁇ g/ml and the scaffold proteins TtSlyDcys-HERl, TgSlyDcys-Herl and TtSlyDcas were immobilized at a concentration of 0.5 ⁇ g/ml.
  • Hybridoma Supernatant was added to the plates and incubated for 1 h at room temperature.
  • HRP Horse radish peroxidase
  • F(ab , ) 2 goat anti-mouse Fey Dianova
  • ABTS 2,2'-Azino-di-[3-ethylbenzthiazoline sulfonate (6)] diammonium salt
  • HRP-labeled F(ab " ) 2 ABTS (Roche) was used as a HRP-substrate.
  • HEK293 cells were transiently transfected with plasmids coding for full length
  • the slides were dewaxed and antigen retrieval was performed by applying heat for 1 hour.
  • the Ventana buffer CC1 was used for antigen retrieval.
  • the antibodies were used at a concentration of 1 ⁇ g/ml.
  • the Ventana Ultra View detection kit was used for the detection of bound antibody. Results are shown in
  • RNA sequences of the selected hybridoma clones a 5 " Race PCR was conducted.
  • total RNA are prepared from 5xl0 6 cells by using a total RNA purification kit (Qiagen).
  • the reverse transcription and the PCR were conducted using a S ' prime RACE PCR kit (Roche).
  • the resulting PCR fragments from heavy and light chain are purified by gel electrophoresis and subsequent gel purification.
  • the PCR fragments are cloned using the Topo Zero-Blunt cloning kit (Invitrogen) and transformed into competent cells.
  • a peptide-based epitope mapping experiment was done to characterize the HER1 ECD epitopes by using the CelluSpotsTM Synthesis and Epitope Mapping technology.
  • Epitope mappings were carried out by means of a library of overlapping, immobilized peptide fragments (length: 15 amino acids) corresponding to the sequences of human HER1 ECD, HER2 ECD, HER3 ECD and HER4 ECD peptide hairpins.
  • Figure 6 the strategy of the epitope mapping is shown.
  • the peptide hairpin sequences ( ⁇ -hairpin) of HERl(EGFR) ECD, HER2 ECD, HER3 ECD and HER4 ECD including their structural embeddings (structural) were investigated.
  • Cysteins were replaced by serines. Each peptide synthesized was shifted by one amino acid, i.e. it had 14 amino acids overlap with the previous and the following peptide, respectively.
  • Intavis CelluSpotsTM technology was employed for preparation of the peptide arrays.
  • peptides are synthesized with an automated synthesizer (Intavis MultiPep RS) on modified cellulose disks which are dissolved after synthesis. The solutions of individual peptides covalently linked to macromolecular cellulose are then spotted onto coated microscope slides.
  • the CelluSpotsTM synthesis was carried out stepwise utilizing 9-fluorenylmethoxycarbonyl (Fmoc) chemistry on amino- modified cellulose disks in a 384-well synthesis plate. In each coupling cycle, the corresponding amino acids were activated with a solution of DIC/HOBt in DMF.
  • Fmoc 9-fluorenylmethoxycarbonyl
  • the peptide solutions were subsequently spotted onto Intavis CelluSpotsTM slides using an Intavis slide spotting robot.
  • TBS Tris-buffered saline
  • the slides prepared as described above were washed with ethanol and then with Tris-buffered saline (TBS; 50 mM Tris, 137 mM NaCl, 2.7 mM KC1, pH 8) before blocking for 16 h at 4°C with 5 mL lOx Western Blocking Reagent (Roche Applied Science), 2.5 g sucrose in TBS, 0.1% Tween 20.
  • the slide was washed with TBS and 0.1% Tween 20 and incubated afterward with 1 ⁇ g/mL of the corresponding IGF1 antibodies in TBS and 0.1% Tween 20 at ambient temperature for 2 h and subsequently washed with TBS + 0.1 % Tween 20.
  • the slide was incubated with anti-rabbit / anti-mouse secondary HRP- antibody (1 :20000 in TBS-T) followed by incubation with chemiluminescence substrate luminol and visualized with a Lumilmager (Roche Applied Science).
  • ELISA-positive SPOTs were quantified and through assignment of the corresponding peptide sequences the antibody binding epitopes were identified.
  • M-47-13 shows a HERl ECD epitope with the amino acid sequence TYQMDVNPEG ( SEQ ID NO: 19) with no detectable signals versus the hairpin motives in the HER2 ECD, the HER3 ECD or the HER4 ECD M31-22 shows a slightly different epitope.
  • M-50-14 deposited MAK ⁇ HER1-DIB> M-
  • the kinetic screening is performed according to Schraeml et al. (Schraml, M. and M. Biehl, Methods Mol Biol 901 (2012) 171-181) on a BIAcore 4000 instrument, mounted with a Biacore CM5 sensor. In all assay the test antibodies are captured. The system is under the control of the software version VI .1.
  • the instrument buffer was HBS-EP (10 mM HEPES (pH 7.4), 150 mM NaCl, 1 mM EDTA, 0.05 % (w/v) P20). The system is operated at 25 °C.
  • the antigen human recombinant FIERI ECD, and one of the recombinant Thermus thermophilus TtSlyDcas-HERl, TtSlyDcys-HERl, TtSlyD(GSG)-HERl, TtSlyD(CC)-HERl, TtSlyD(SS)-HERl, Thermococcus gammatolerans TgSlyDcys-HERl comprising the ⁇ -hairpin peptide of FIERI (SEQ ID NO: l) ) is diluted at 150 nM in instrument buffer supplemented with lmg/ml CMD(Carboxymethyldextran, Sigma), to suppress unspecific binding.
  • FIERI ECD human recombinant FIERI ECD, and one of the recombinant Thermus thermophilus TtSlyDcas-HERl, TtSlyDcys-HERl, TtSlyD(GSG)-HERl, TtSlyD
  • the hybridoma culture supernatants Prior to their application the hybridoma culture supernatants are diluted 1 :5 in instrument buffer. The diluted mixtures are injected at a flow rate of 30 ⁇ /min for 2 min. The antibody capture level (CL) in response units is monitored. Immediately thereafter the respective antigen is injected at a flow rate of 30 ⁇ /min for 3 min association time. Thereafter, the antibody-antigen complex dissociation signal is recorded for 5 min. The sensor is regenerated by injecting a 10 mM glycine-HCl solution (pH 1.7) for 2 min at a flow rate of 30 ⁇ /min. The recorded signal shortly before the end of the injection of the antigen is denoted as binding late (BL) in response units.
  • BL binding late
  • the recorded signal shortly before the end of the recording of the dissociation is denoted as stability late (SL) in response units.
  • the dissociation rate constants are determined calculated.
  • the antibody-antigen complex stability in minutes is calculated with the following formula: ln(2)/60*kd .
  • the Molar Ratio was calculated with the formula: MW (antibody) / MW( antigen) *BL (antigen)/ CL
  • Binding Late represents the response units at the end of the analyte injection.
  • the amount of antibody captured as a ligand on the sensor surface is measured as Capture Level (CL) in response units.
  • CL Capture Level
  • the Molar Ratio can be calculated.
  • the Molar Ratio can indicate understoichiometric binding, like it is the case when the HERl ECD is being bound in its "closed" conformation by the anti HERl ⁇ -hairpin antibodies of the invention (as this ⁇ hairpin is hidden in the closed conformation.
  • Binding of anti-HERl ⁇ -hairpin antibodies to and internalization of anti-HERl ⁇ - hairpin antibodies was analyzed in Western Blot using the HERl expressing cancer cell line A549.
  • HER1 expressing A549 (ATCC ® CCL-185TM lung carcinoma) cells and A431 (ATCC ® CRL-1555TM - skin cancer/epidermoid carcinoma) cells were seeded into 24-well-plates (3x105 cells/well, media containing 10% FCS). On the next day the media was replaced by starving media (0.5%> FCS). Four hours later (24 hours before cell lysis), antibody was added to two wells of each cell line to a final concentration of 185 ⁇ g/ml. On the next day antibody was added again at the following times: cell lysis minus 6 hours, minus 4 hours, minus 2 hours, minus 1 hour. Ten minutes before cell lysis one well of each time point was stimulated with hEGF (final concentration 200ng/ml). The cells were lysed with 40 ⁇ 1 Triton Lysis
  • Results are shown in Figure 8A (A549 cells) and 8B (A431 cells, strong HER1 expression with slight constitutively activated/phosphorylated HER1). Left lane shows HER1 detection, right lane the phosphorylated HER1 detection in th absence or presence of EGF
  • HER1 dib3 M-47-15 show no induction of phosphorylation and act as non- agonistic HER1 antibodies (in the absence of EGF ligand, -lane), while other HER1 antibodies like cetuximab, GA201 (imgatuzumab, CAS number 959963-46- 3 a humanized, glycoengineered IgGl mAb derived by humanization of the parental ICR62 rat antibody, described e.g.
  • ABT806 targets the EGFR deletion variant, de2-7 EGFR as well as wild-type, described e.g in US2011/0076232) were induced strong phosphorylation ( in the absence of EGF , compared to medium without antibody or to the antibodies of the present invention).
  • a Streptavidin-coated 96-well plate was incubated at 4°C with cell culture supernatant containing SBP-tagged HER1-ECD. On the next day the wells were washed three times with washing buffer (PBS + 0.05% Tween-20) and blocked with PBS containing 1% BSA for one hour. After another three washes with washing buffer, 40 ⁇ 1 antibody solution (in Delfia Binding Buffer) was added to each well as a 2x stock of the desired final concentrations (10 ⁇ 3 to 10 3 nM,). Immediately 40 ⁇ 1 of 20nM Europium-labeled EGF was added to achieve a final concentration of ⁇ . The plates were incubated on a shaker at room temperature for two hours.
  • Delfia Enhancement Solution was added and incubated on a shaker for 15 minutes (light protected). Finally, the plates were measured in a Tecan Infinite F200 reader using a time -resolved fluorescence measurement protocol. The HERldib-supernatants were used in dilutions from 1 : 1 to l : 10e7.

Abstract

L'invention concerne des protéines de liaison à l'antigène anti-HER1, par exemple des anticorps anti-HER1, qui se lient à l'épingle à cheveux β de HER1, des procédés de sélection de ces protéines de liaison à l'antigène, leur préparation et leur utilisation en tant que médicament.
EP15721718.3A 2014-05-14 2015-05-12 Protéines de liaison d'antigène her1 pour la liaison en épingle à cheveux bêta de her1 Active EP3143046B1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP14168320 2014-05-14
PCT/EP2015/060491 WO2015173249A1 (fr) 2014-05-14 2015-05-12 Protéines de liaison à l'antigène her1 se liant à l'épingle à cheveux beta de her1

Publications (2)

Publication Number Publication Date
EP3143046A1 true EP3143046A1 (fr) 2017-03-22
EP3143046B1 EP3143046B1 (fr) 2020-06-17

Family

ID=50729372

Family Applications (1)

Application Number Title Priority Date Filing Date
EP15721718.3A Active EP3143046B1 (fr) 2014-05-14 2015-05-12 Protéines de liaison d'antigène her1 pour la liaison en épingle à cheveux bêta de her1

Country Status (11)

Country Link
US (2) US9840565B2 (fr)
EP (1) EP3143046B1 (fr)
JP (1) JP6559709B2 (fr)
KR (1) KR20160145119A (fr)
CN (1) CN106459197B (fr)
AR (1) AR100427A1 (fr)
BR (1) BR112016024742A2 (fr)
CA (1) CA2945066A1 (fr)
MX (1) MX2016014726A (fr)
TW (1) TW201605903A (fr)
WO (1) WO2015173249A1 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112016025056A2 (pt) 2014-05-14 2018-02-20 F. Hoffmann-La Roche Ag uso de pelo menos um polipeptídeo, anticorpo biespecífico, anticorpo biespecífico isolado, anticorpo her3/her2 biespecífico, célula hospedeira, método de produção do anticorpo biespecífico her3/her2, imunoconjugado e formulação farmacêutica
US11124575B2 (en) 2017-02-22 2021-09-21 University Of Saskatchewan EGFR-binding agents and uses thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7608413B1 (en) * 2005-03-25 2009-10-27 Celera Corporation Kidney disease targets and uses thereof
JP2009532393A (ja) * 2006-04-07 2009-09-10 コペンハーゲン ユニバーシティ ErbB受容体由来ペプチド断片
AR086250A1 (es) * 2011-05-05 2013-11-27 Hoffmann La Roche Polipeptido de fusion presentador de una secuencia de aminoacidos y utilizacion del mismo

Also Published As

Publication number Publication date
US9840565B2 (en) 2017-12-12
BR112016024742A2 (pt) 2018-01-30
CA2945066A1 (fr) 2015-11-19
AR100427A1 (es) 2016-10-05
JP2017523391A (ja) 2017-08-17
JP6559709B2 (ja) 2019-08-14
US20180100022A1 (en) 2018-04-12
CN106459197B (zh) 2021-01-08
TW201605903A (zh) 2016-02-16
EP3143046B1 (fr) 2020-06-17
WO2015173249A1 (fr) 2015-11-19
KR20160145119A (ko) 2016-12-19
MX2016014726A (es) 2017-03-23
US20160002346A1 (en) 2016-01-07
CN106459197A (zh) 2017-02-22

Similar Documents

Publication Publication Date Title
US10808038B2 (en) HER3/HER2 bispecific antibodies binding to the beta-hairpin of HER3 and domain II of HER2
US10626177B2 (en) HER3 antigen binding proteins binding to the beta-hairpin of HER3
US10364290B2 (en) Anti-HER3/HER4 antibodies binding to the beta-hairpin of HER3 and the beta-hairpin of HER4
US9783611B2 (en) Anti-HER3 antibodies binding to the beta-hairpin of HER3
US20180100022A1 (en) Her1 antigen binding proteins binding to the beta-hairpin of her1

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20161214

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

RIN1 Information on inventor provided before grant (corrected)

Inventor name: SCHRAEML, MICHAEL

Inventor name: BOSSENMAIER, BIRGIT

Inventor name: NIEDERFELLNER, GERHARD

Inventor name: GERG, MICHAEL

Inventor name: PEESS, CARMEN

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20180119

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20200207

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602015054333

Country of ref document: DE

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1281189

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200715

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200917

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200918

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20200617

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200917

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1281189

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200617

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201019

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201017

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602015054333

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

26N No opposition filed

Effective date: 20210318

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20210413

Year of fee payment: 7

Ref country code: FR

Payment date: 20210420

Year of fee payment: 7

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20210412

Year of fee payment: 7

Ref country code: GB

Payment date: 20210428

Year of fee payment: 7

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210512

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20210531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210512

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210531

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602015054333

Country of ref document: DE

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20220512

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220531

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20150512

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20220512

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221201

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200617